WO2015108992A1 - Heterobicyclic compounds and their use as fgfr4 receptor inhibitors - Google Patents

Heterobicyclic compounds and their use as fgfr4 receptor inhibitors Download PDF

Info

Publication number
WO2015108992A1
WO2015108992A1 PCT/US2015/011424 US2015011424W WO2015108992A1 WO 2015108992 A1 WO2015108992 A1 WO 2015108992A1 US 2015011424 W US2015011424 W US 2015011424W WO 2015108992 A1 WO2015108992 A1 WO 2015108992A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
fgfr
treating
optionally substituted
alkyl
Prior art date
Application number
PCT/US2015/011424
Other languages
French (fr)
Inventor
Neil Bifulco, Jr.
Chandrasekhar V. MIDUTURU
Lucian V. Dipietro
Original Assignee
Blueprint Medicines Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Blueprint Medicines Corporation filed Critical Blueprint Medicines Corporation
Publication of WO2015108992A1 publication Critical patent/WO2015108992A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/12Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring
    • C07D217/14Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/12Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring
    • C07D217/14Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals
    • C07D217/16Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/74Quinazolines; Hydrogenated quinazolines with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, attached to ring carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems

Definitions

  • Described herein are compounds, pharmaceutical compositions, and methods of using such compounds and compositions to inhibit the activity of FGFR-4.
  • Fibroblast growth factor receptor 4 is a protein that in humans is encoded by the FGFR-4 gene. This protein is a member of the fibroblast growth factor receptor family, where amino acid sequence was highly conserved between members throughout evolution. FGFR family members 1-4 differ from one another in their ligand affinities and tissue distribution. A full-length representative protein consists of an extracellular region composed of three immunoglobulin-like domains, a single hydrophobic membrane-spanning segment and a cytoplasmic tyrosine kinase domain. The extracellular portion of the protein interacts with fibroblast growth factors, setting in motion a cascade of downstream signals, ultimately influencing mitogenesis and differentiation. The genomic organization of the FGFR-4 gene encompasses 18 exons. Although alternative splicing has been observed, there is no evidence that the C-terminal half of the Iglll domain of this protein varies between three alternate forms, as indicated for FGFR 1-3.
  • inhibitors of FGFR-4 are inhibitors of FGFR-4. Further described herein are pharmaceutical compositions and pharmaceutical formulations that include an inhibitor of FGFR-4.
  • the invention features a compound of Formula I, or pharmaceutically acceptable salt thereof, wherein:
  • Formula I denotes a single or double bond
  • Warhead is a moiety capable of forming a covalent bond with a nucleophile
  • Ring A is a 5-8 membered aryl, 5-12 membered heteroaryl, 3-7 member heterocyclyl or 3-12 membered cycloalkyl group;
  • W is C or N , X and Z are each independently CH or N;
  • Y is CH or N-R 4 wherein R 4 is H, Ci_6 alkyl, or 3-12 membered cycloalkyl;
  • each of R 1 -R 3 is, independently, halo, cyano, optionally substituted Ci_6 alkyl, optionally substituted Ci_6 alkoxy, hydroxy, oxo, amino, amido, alkyl urea, optionally substituted 3-7 member heterocyclyl;
  • R 7 is hydrogen or Ci alkyl
  • Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R 1 ;
  • n 0-5;
  • n 0-5;
  • p 0-2.
  • a warhead is a moiety that is reactive with a nucleophile, for example, capable of forming a covalent bond with a nucleophile.
  • warheads include, without limitation, alkyl halides, alkyl sulfonates, heteroaryl halides, epoxides, haloacetamides, maleimides, sulfonate esters, alpha-beta unsaturated ketones, alpha-beta unsaturated esters, vinyl sulfones, propargyl amides, acrylamides.
  • the N of the warhead is the adjacent N in the formulae shown above. Structures of exemplary warheads are shown below:
  • X is a leaving group such as halo, or an activated hydroxyl moiety (e.g. , triflate); and each of R a , R b , and R c is, independently, H, substituted or unsubstituted C 1-4 alkyl, substituted or unsubstituted C 1-4 cycloalkyl, or cyano.
  • the warheads are typically attached to a N atom on the inhibitor.
  • the warhead can alternatively be attached to an atom other than N.
  • Examples of exemplary warheads include, without limitation,
  • warheads can be found, e.g. , in WO 2010/028236 and WO 2011/034907.
  • the invention features a method for treating a condition mediated by FGFR-4, a condition characterized by overexpression of FGFR-4, a condition characterized by amplification of FGFR4, a condition mediated by FGF19, a condition characterized by amplified FGF- 19, or a condition characterized by overexpression of FGF19, any of these methods comprising administering a therapeutically effective amount of a compound disclosed herein to a subject.
  • the invention features a method of treating any of the following conditions by administering a therapeutically effective amount of a compound disclosed herein to a subject: hepatocellular carcinoma, breast cancer, ovarian cancer, lung cancer, liver cancer, a sarcoma, or hyperlipidemia.
  • the invention includes all possible combinations of the embodiments described above and below.
  • the compounds disclosed herein can form a covalent bond with FGFR-4 protein; for example, the compounds can form a covalent bond with a cysteine residue of FGFR-4, for example, the cysteine at residue 552. FGFRsl-3 do not contain this cysteine.
  • the ability to form a covalent bond between the compound and FGFR-4 is therefore an important factor in the selectivity of the compounds disclosed herein for FGFR-4.
  • Aliphatic group refers to a straight-chain, branched-chain, or cyclic hydrocarbon group and includes saturated and unsaturated groups, such as an alkyl group, an alkenyl group, and an alkynyl group.
  • Alkenyl refers to an aliphatic group containing at least one double bond.
  • Alkoxyl or "alkoxy”, as used herein, refers to an alkyl group having an oxygen radical attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
  • Alkyl refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl- substituted cycloalkyl groups, and cycloalkyl- substituted alkyl groups.
  • Alkylene refers to a double radical, that is, an aliphatic group substituted on two ends.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g. , Q-C 30 for straight chains, C 3 -C 30 for branched chains), and in other embodiments can have 20 or fewer, or 10 or fewer.
  • cycloalkyls may have from 3- 10 carbon atoms in their ring structure, and in some embodiments may have 5, 6 or 7 carbons in the ring structure.
  • alkenyl refers to an aliphatic group containing at least one double bond
  • alkynyl refers to an aliphatic group containing at least one triple bond.
  • Alkylthio refers to a hydrocarbyl group having a sulfur radical attached thereto.
  • the "alkylthio" moiety is represented by one of -S-alkyl, -S- alkenyl, or -S-alkynyl.
  • Representative alkylthio groups include methylthio, ethylthio, and the like.
  • R and R is independently hydrogen, hydroxyl, C C 6 alkyl, or C C 6 alkoxyl, e.g. , methoxyl.
  • Amino refers to -NH 2 , -NH(alkyl), or -N(alkyl) (alkyl).
  • Aminified means additional copies of a gene or chromosome segment are produced in cancer cells that may confer a growth or survival advantage.
  • Alkyl refers to an alkyl group substituted with an aryl group (e.g. , an aromatic or heteroaromatic group).
  • Aryl refers to 5-, 6-, and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, phenyl, pyrrolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyrazolyl, pyridinyl, pyrazinyl, pyridazinyl and pyrimidinyl, and the like.
  • aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles" or “heteroaromatics.”
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, polycyclyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are "fused rings") wherein at least one of the rings is aromatic, e.g. , the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls. Each ring can contain, e.g. , 5-7 members.
  • carrier refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon.
  • cycloalkyl as used herein, includes cyclic, bicyclic, tricyclic, or polycyclic non-aromatic hydrocarbon groups having 3 to 12 carbons. Any substitutable ring atom can be substituted (e.g. , by one or more substituents).
  • the cycloalkyl groups can contain fused or spiro rings. Fused rings are rings that share a common carbon atom. Examples of cycloalkyl moieties include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl,
  • Covalent inhibitor means an inhibitor that can form a covalent bond with a protein.
  • enantiomeric excess or "% enantiomeric excess” of a composition can be calculated using the equation shown below.
  • a composition contains 90% of one enantiomer, e.g., the S-enantiomer, and 10% of the other enantiomer, i.e., the R- enantiomer.
  • compositions containing 90% of one enantiomer and 10% of the other enantiomer is said to have an enantiomeric excess of 80%.
  • the compositions described herein contain an enantiomeric excess of at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the S-enantiomer.
  • the compositions contain an enantiomeric excess of the S-enantiomer over the R-enantiomer.
  • the compositions described herein contain an enantiomeric excess of at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the R- enantiomer. In other words, the compositions contain an enantiomeric excess of the R- enantiomer over the S-enantiomer.
  • FGFR-4" or "FGFR-4 protein” refers to any form of the FGFR-4 protein, including wild type and all variant forms (including, without limitation, mutant forms and splice variants).
  • the FGFR-4 protein is a product of the FGFR-4 gene, and the FGFR-4 protein therefore includes any protein encoded by any form of the FGFR-4 gene, including all aberrations, e.g., point mutations, indels, translocation fusions, and focal amplifications.
  • Halo refers to a radical of any halogen, e.g., -F, -CI, -Br, or -I.
  • Haloalkyl and haloalkoxy refers to alkyl and alkoxy structures that are substituted with one or more halo groups or with combinations thereof.
  • fluoroalkyl and fluoro alkoxy include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine.
  • Heteroarylalkyl refers to an alkyl group substituted with a heteroaryl group.
  • Heterocyclyl or “heterocyclic group” refers to a ring structure, such as a 3- to 14- membered ring structure, whose ring(s) include one or more heteroatoms. Heterocyclyl can also include polycycles, with each group having, e.g., 3-7 ring members.
  • saturated or partially saturated heterocyclyl refers to a non-aromatic cyclic structure that includes at least one heteroatom.
  • the term “heterocyclyl” can also include 8-12 membered bicyclic heterocyclyls, e.g., wherein a saturated or partially saturated
  • heterocyclyl is fused to an aromatic or heteroaromatic ring.
  • heterocyclyl can also include 8- 12 membered bicyclic heterocyclyls, e.g. , wherein a saturated or partially saturated cycloalkyl is fused to an aromatic or heteroaromatic ring.
  • the point of attachment of the heterocyclyl to the rest of the molecule can be through the saturated or partially saturated heterocyclyl or cycloalkyl, or through the aromatic or heteroaromatic ring.
  • Heteroaryl refers to a 5-14 membered (i.e. , 5-8 membered monocyclic, 8- 12 membered bicyclic, or 11-14 membered tricyclic) aromatic ring system having 1-3 ring heteroatoms if monocyclic, 1-6 ring heteroatoms if bicyclic, or 1-9 ring heteroatoms if tricyclic, said ring heteroatoms independently selected from O, N, and S (e.g. , 1-3, 1-6, or 1-9 ring heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively). Any ring atom can be substituted (e.g. , by one or more substituents).
  • Heterocyclyl groups include, for example, thiophenyl, thianthrenyl, furanyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathiin, pyrrolyl, imidazolyl, pyrazolyl, isothiazolyl, isoxazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, quinolizinyl, isoquinolinyl, quinolinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, quinazolinyl, cinnolinyl, pteridinyl, carbazolyl, carboline, phenanthridine, acridine, pyrimidine, phen
  • the heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF 3 , - CN, or the like.
  • substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phos
  • Heterocyclylalkyl refers to an alkyl group substituted with a heterocycle group.
  • an inhibitor refers to a compound that inhibits an enzyme such that a reduction in activity of the enzyme can be observed, e.g. , in a biochemical assay.
  • an inhibitor has an IC 50 of less than about 1 ⁇ , less than about 500 nM, less than about 250 nM, less than about 100 nM, less than about 50 nM, or less than about 10 nM.
  • An inhibitor of FGFR- 4 refers to a compound that inhibits FGFR-4.
  • Nucleophile refers to a species that donates an electron-pair to an electrophile to form a chemical bond in a reaction.
  • a nucleophile can be an oxygen nucleophile, e.g.
  • a nitrogen nucleophile e.g. , amine
  • a sulfur nucleophile e.g. , thiol
  • thiol such as, for example, the thiol in the side chain of a cysteine residue.
  • “Overexpressed,” as used herein, means there is production of a gene product in a sample that is substantially higher than that observed in a population of control samples (e.g. normal tissue).
  • FGFR- 4 refers to a compound that inhibits the activity of a target protein, e.g. , FGFR- 4, more potently than it inhibits activity of other proteins.
  • the isoforms FGFR- 1, FGFR-2, FGFR-3, and FGFR-4 are all considered distinct proteins.
  • a compound can inhibit the activity of the target protein, e.g. , FGFR-4, at least 1.5, at least 2, at least 5, at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 200, at least 500, or at least 1000 or more times potently than it inhibits the activity of a non-target protein.
  • Substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g. , which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic mo
  • the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate.
  • the substituents of a substituted alkyl may include substituted and unsubstituted forms of amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), -CF 3 , -CN and the like.
  • Cycloalkyls can be further substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, -CF 3 , -CN, and the like.
  • Analogous substitutions can be made to alkenyl and alkynyl groups to produce, for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls, iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls.
  • each expression e.g. , alkyl, m, n, etc., when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
  • Warhead moiety refers to a moiety of an inhibitor which participates, either reversibly or irreversibly, with the reaction of a donor, e.g. , a protein, with a substrate.
  • Warheads may, for example, form covalent bonds with the protein, or may create stable transition states, or be a reversible or an irreversible alkylating agent.
  • the warhead moiety can be a functional group on an inhibitor that can participate in a bond-forming reaction, wherein a new covalent bond is formed between a portion of the warhead and a donor, for example an amino acid residue of a protein.
  • the warhead is an electrophile and the "donor" is a nucleophile such as the side chain of a cysteine residue.
  • suitable warheads include, without limitation, the groups shown below:
  • X is a leaving group such as halo, or an activated hydroxyl moiety (e.g. , triflate); and each of R a , R b , and R c is, independently, H, substituted or unsubstituted C 1-4 alkyl, substituted or unsubstituted Ci_4 cycloalkyl, or cyano.
  • the term "patient” or “subject” refers to organisms to be treated by the methods of the present invention.
  • Such organisms preferably include, but are not limited to, mammals (e.g. , murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans.
  • the term "effective amount” refers to the amount of a compound (e.g. , a compound of the present invention) sufficient to effect beneficial or desired results.
  • An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route.
  • the term “treating” includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
  • the term “modulate” or “modulating” refers to an increase or decrease, e.g. , in the activity of an enzyme in response to exposure to a compound or composition described herein, e.g. , the inhibition of FGFR-4, in at least a sub-population of cells in a subject such that a desired end result is achieved (e.g. , a therapeutic result).
  • a compound as described herein inhibits a target described herein, e.g. , FGFR-4.
  • the compounds described herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H) or carbon- 14 ( 14 C). All isotopic variations of the compounds disclosed herein, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
  • deuterated compounds such as for example tritium ( 3 H) or carbon- 14 ( 14 C).
  • the compounds described herein can be useful as the free base or as a salt.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like.
  • sulfate bisulfate
  • phosphate nitrate
  • acetate valerate
  • oleate palmitate
  • stearate laurate
  • benzoate lactate
  • phosphate tosylate
  • citrate maleate
  • fumarate succinate
  • tartrate napthylate
  • mesylate glucoheptonate
  • lactobionate lactobionate
  • laurylsulphonate salts and the like See, for example,
  • Certain compounds disclosed herein can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention. Certain compounds disclosed herein may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • the invention provides a compound having structural Formula I:
  • Warhead is a moiety capable of forming a covalent bond with a nucleophile
  • Ring A is a 5-8 membered aryl, 5-12 membered heteroaryl, 3-7 member heterocyclyl or 3-12 membered cycloalkyl group;
  • W is C or N, X and Z are each independently CH or N;
  • Y is CH or N-R 4 where R 4 is H, C 1-6 alkyl, or 3-12 membered cycloalkyl;
  • each of R 1 -R 3 is, independently, halo, cyano, optionally substituted C 1-6 alkyl, optionally substituted Ci_6 alkoxy, hydroxy, oxo, amino, amido, alkyl urea, optionally substituted 3-7 member heterocyclyl;
  • R 7 is hydrogen or C 1-6 alkyl; or R 7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R 1 ;
  • n 0-5;
  • n 0-5;
  • p 0-2.
  • Ring A is aryl. In some embodiments, Ring A is phenyl substituted with 0-3 R 1 . In some embodiments, R 1 is selected from the group consisting of halo, optionally substituted C 1-6 alkyl, and optionally substituted C 1-6 alkoxy.
  • R 7 is hydrogen or Ci_6 alkyl. In some embodiments, R 7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R 1 1 . In some embodiments, R 7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-3 occurrences of R 1 .
  • X is N. In some embodiments, X is CH.
  • Y is N-R 4 wherein R 4 is Ci_6 alkyl or 3-12 membered cycloalkyl.
  • R 4 is methyl, ethyl or isopropyl.
  • R 4 is ethyl.
  • R 4 is 3-12 membered cycloalkyl.
  • R 4 is cyclopentyl.
  • Y is N-R 4 wherein R 4 is Ci_6 alkyl or 3-12 membered cycloalkyl and R 3 is oxo.
  • R 4 is methyl, ethyl or isopropyl.
  • R 4 is ethyl.
  • R 4 is 3-12 membered cycloalkyl.
  • R 4 is cyclopentyl.
  • q is 0.
  • the warhead moiety is selected from the group consisting of
  • each of R a , R , and R c is, independently, hydrogen, optionally substituted C 1-4 alkyl, optionally substituted 3-12 membered cycloalkyl, or cyano.
  • R 2 is alkoxy or halo. In some embodiments, R 2 is methoxy or chloro. In some embodiments, two R 2 are chloro and two R 2 are methoxy.
  • the invention features a compound of Formula 1(a) or a
  • X is CH or N. In some embodiments, X is CH. In some embodiments, X is
  • the invention features a compound of Formula 1(b) or a
  • X is CH or N. In some embodiments, X is CH. In some embodiments, X is
  • the invention features a compound of Formula 1(b) or a pharmaceutically acceptable salt thereof:
  • X is CH or N. In some embodiments, X is CH. In some embodiments, X is
  • the invention features a compound of Formula 1(d) or a pharmaceutically acce table salt thereof:
  • the Warhead moiety is Ring A is phenyl, and R 7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R ;
  • R is C 1-6 alkyl or 3-12 membered cycloalkyl;
  • R is Ci_6 alkoxy or halo; and
  • n is 4.
  • R together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-3 occurrences of R 1 .
  • Ring A together with R forms an 8-10 membered heterocyclyl. In some embodiments, Ring A together with R forms:
  • V is CH 2 or O; r is 0 or 1; and R 1 and m are as defined herein.
  • Ring A together with R forms:
  • V is CH 2 or O; r is 0 or 1 ; and R and m are as defined herein.
  • the invention also features pharmaceutical compositions comprising a pharmaceutically acceptable carrier and any compound of Formulas 1, 1(a), 1(b), 1(c), and 1(d).
  • a compound disclosed herein While it is possible for a compound disclosed herein to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation, where the compound is combined with one or more pharmaceutically acceptable excipients or carriers.
  • the compounds disclosed herein may be formulated for administration in any convenient way for use in human or veterinary medicine.
  • the compound included in the pharmaceutical preparation may be active itself, or may be a prodrug, e.g. , capable of being converted to an active compound in a physiological setting.
  • the compounds provided herein include their hydrates.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • Examples of pharmaceutically acceptable salts of a compound described herein include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • Suitable acid salts include acetate, adipate, benzoate, benzenesulfonate, butyrate, citrate, digluconate, dodecylsulfate, formate, fumarate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactate, maleate, malonate,
  • salts derived from appropriate bases include alkali metal (e.g. , sodium), alkaline earth metal (e.g. , magnesium), ammonium and N-(alkyl) 4 + salts.
  • alkali metal e.g. , sodium
  • alkaline earth metal e.g. , magnesium
  • ammonium e.g. 0.5
  • Examples of pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) algin
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium
  • antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like
  • metal chelating agents such as citric acid
  • Solid dosage forms can include one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6)
  • Liquid dosage forms can include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such
  • Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
  • the amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect.
  • Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required.
  • the compounds disclosed herein are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • the formulations can be administered topically, orally, transdermally, rectally, vaginally, parentally, intranasally, intrapulmonary, intraocularly, intravenously, intramuscularly, intraarterially, intrathecally, intracapsularly, intradermally, intraperitoneally, subcutaneously, subcuticularly, or by inhalation.
  • FGFR-4 regulates proliferation, survival, and alpha-fetoprotein secretion during hepatocellular carcinoma (HCC) progression; inhibitors of FGFR-4 are therefore promising potential therapeutic agents for this unmet medical need (Ho et al., Journal of Hepatology, 2009, 50: 118-27). HCC afflicts more than 550,000 people worldwide every year and has one of the worst 1-year survival rates of any cancer type.
  • FGF 19 fibroblast growth factor (FGF) family, which consists of hormones that regulate glucose, lipid, and energy homeostasis. Increased hepatocyte
  • FGF 19 activates FGFR-4, its predominant receptor in the liver, and it is believed that activation of FGFR-4 is the mechanism whereby FGF 19 can increase hepatocyte proliferation and induce hepatocellular carcinoma formation (Wu et al., J Biol Chem (2010) 285(8):5165-5170). FGF 19 has been identified as a driver gene in HCC by others as well (Sawey et al., Cancer Cell (2011) 19: 347-358). It is therefore believed that the compounds disclosed herein, which are potent and selective inhibitors of FGFR-4, can be used to treat HCC and other liver cancers.
  • Oncogenome screening has identified an activating fibroblast growth factor receptor 4 (FGFR-4) Y367C mutation in the human breast cancer cell line MDA-MB-453. This mutation was shown to elicit constitutive phosphorylation, leading to an activation of the mitogen- activated protein kinase cascade. Accordingly, it has been suggested that FGFR-4 may be a driver of tumor growth in breast cancer (Roidl et al., Oncogene (2010) 29(10): 1543-1552). It is therefore believed that the compounds disclosed herein, which are potent and selective inhibitors of FGFR-4, can be used to treat FGFR-4 modulated breast cancer.
  • FGFR-4 fibroblast growth factor receptor 4
  • FGFR-4 Molecular changes (e.g. , translocations) in genes upstream of FGFR-4 can lead to activation/overexpression of FGFR-4.
  • a PAX3-FKHR translocation/gene fusion can lead to FGFR-4 overexpression.
  • Overexpression of FGFR-4 due to this mechanism has been associated with rhabdomyosarcoma (RMS) (Cao et al., Cancer Res (2010) 70(16): 6497-6508).
  • RMS rhabdomyosarcoma
  • Mutations in FGFR-4 itself can lead to over-activation of the protein; this mechanism has been associated with a subpopulation of RMS (Taylor et al., J Clin
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound disclosed herein employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a compound of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • FGFR-4 inhibitors disclosed herein can be combined with other cancer treatments.
  • the inhibitors can be administered in combination with surgical treatments, radiation, or other therapeutic agents such as antibodies, other selective kinase inhibitors, or chemotherapeutics.
  • the inhibitors may also be administered in combination with RNAi therapy or antisense therapy.
  • the FGFR-4 inhibitors described herein may be combined with one, two, or more other therapeutic agents. Examples of other therapeutic agents include sorafenib, gemcitabine, capecitabine, and doxorubicin.
  • second therapeutic agent also includes more than one therapeutic agent other than the FGFR-4 inhibitor.
  • a FGFR-4 inhibitor described herein may be administered with one, two, or more other therapeutic agents.
  • the FGFR-4 inhibitors described herein and the second therapeutic agent do not have to be administered in the same pharmaceutical composition, and may, because of different physical and chemical characteristics, be administered by different routes.
  • the FGFR-4 inhibitor can be administered orally, while the second therapeutic agent is administered intravenously.
  • the determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician.
  • the initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
  • the FGFR-4 inhibitor and the second therapeutic agent may be administered
  • FGFR-4 inhibitors disclosed herein can be administered as part of an antibody-drug conjugate, where the FGFR-4 inhibitor is the "payload" portion of the conjugate.
  • the table below shows exemplary compounds.
  • Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g. , temperatures which can range from the solvent' s freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by the skilled artisan.
  • Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4th ed., John Wiley & Sons: New Jersey, (2006), which is incorporated herein by reference in its entirety.
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic
  • NMR nuclear magnetic resonance
  • IR infrared
  • MS mass spectrometry
  • chromatographic methods such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC).
  • LCMS Liquid chromatography-mass spectrometry (LCMS) data (sample analyzed for purity and identity) can be obtained with an Agilent model- 1260 LC system using an Agilent model 6120 mass spectrometer utilizing ES-API ionization fitted with an Agilent Poroshel 120 (EC-C18, 2.7um particle size, 3.0 x 50mm dimensions) reverse-phase column at 22.4 degrees Celsius.
  • the mobile phase can be of a mixture of solvent 0.1% formic acid in water and 0.1% formic acid in acetonitrile. A constant gradient from 95% aqueous/5% organic to 5%
  • aqueous/95% organic mobile phase over the course of 4 minutes can be utilized.
  • Step 5 S nthesis of 2-chloro-6-(3,5-dimethox henyl)quinazoline
  • Step 1 Synthesis of eth l 6-chloro-4-(methylamino)nicotinate
  • Step 2 Synthesis of 6-chloro-4-(methylamino)pyridin-3-yl)methanol
  • ethyl 6-chloro-4-(methylamino)nicotinate 4.7 g, 21.9 mmol
  • lithium borohydride 2.4 g, 109.8 mmol
  • the reaction was heated at 55 °C overnight.
  • LCMS showed the reaction was completed.
  • the reaction was cooled to RT, quenched with water (1 mL) and filtered. The filtrate was concentrated to afford the title compound (4.2 g, crude) as a white solid, which was directly used in the next step without further purification.
  • MS (ES+) C 7 H 9 C1N 2 0 requires: 172, 174, found: 173, 175 [M + H] + .
  • Step 3 Synthesis o -chloro-4-(methylamino)nicotinaldehyd
  • Step 4 Synthesis of 7-chloro-3-(3,5-dimethoxyphenyl)-l-methyl-l,6-naphthyridin-2(lH)-one
  • Step 5 Synthesis of 7-chloro-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl-l,6-naphthyridin- 2(lH)-one
  • Step 1 Synthesis of 7-chloro-l-ethyl-3-(2-fluoro-3,5-dimethoxyphenyl)-l,6-naphthyridin-2(lH)- one
  • Step 2 Synthesis of 7-chloro-3-(2-chloro-6-fluoro-3,5-dimethoxyphenyl)-l-ethyl-l,6- naphthyridin-2(lH)-one
  • Step 1 Synthesis of ethyl 6-chloro-4-(ethylamino)nicotinate
  • Step 3 Synthesis of 6-chloro-4-(ethylamino)nicotinaldehyde (Notebook: SP-0010571-035):
  • Step 4 Synthesis of ethyl 2-chloro-5-((3,5-dimethoxyphenylamino)methyl)-N-ethylpyridin-4- amine
  • Step 5 Synthesis of 7-chloro-3-(3,5-dimethoxyphenyl)- l-ethyl-3,4-dihydropyrido[4,3- d]pyrimidin-2(lH)-one
  • Step 6 Synthesis of 7-chloro-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-ethyl-3,4- dihydropyrido[4,3-d]pyrimidin-2(lH)-one
  • Step 1 Synthesis of eth l 4-(methylamino)-2-(methylthio)pyrimidine-5-carboxylate
  • Step 5 Synthesis of 6-(3,5-dimethoxyphenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin- 7(8H)-o
  • Step 6 Synthesis of 6-(3,5-dimethoxyphenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3- d]pyrimidin-7(8H)-one
  • Step 7 Synthesis of 6-(2,6-dichloro-3,5-dimethoxyphenyl)-8-methyl-2- (methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one
  • Step 1 Synthesis of 6-bromo-7-fluoro- -dimethyl-2H-benzo[b][l,4]oxazin-3(4H)-one
  • 2-amino-4-bromo-5-fluorophenol hydrochloride (1.0 g, 4.12 mmol) was taken up in dichloroethane (10 ml) and 2-bromo-2-methylpropanoyl bromide (0.692 ml, 4.54 mmol) was added, followed by addition of DIEA (0.720 ml, 4.12 mmol). Stirred at room temperature for 3 hours. Reaction was cooled to OoC and water added, stirred at OoC for 10 minutes. Layers were separated and solvent removed. Residue was then taken back up in acetonitrile and K2C03(1.65 g, 12 mmol) added. Reaction was then heated to reflux overnight. Complete conversion to desired product.
  • Step 3 Synthesis of 7-fluoro-2,2-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4- dihydro-2H-benzo[b] [ 1 ,4] oxazine
  • 6-bromo-7-fluoro-2,2-dimethyl-3,4-dihydro-2H-benzo[b][l,4]oxazine (0.210 g, 0.807 mmol)
  • BIS(PINACOLATO)DIBORON (0.410 g, 1.615 mmol
  • PdC12(dppf)-CH2C12Adduct 0.066 g, 0.081 mmol
  • POTASSIUM ACETATE 0.238 g, 2.422 mmol
  • Step 3 Synthesis of 6-bromo-7-methyl-3,4-dihydro-2H-benzo[b][l,4]oxazine 6-Bromo-7-methyl-2H-benzo[b][l,4]oxazin-3(4H)-one (84.6 mg, 0.349 mmol) was stirred at 0°C in tetrahydrofuran (2 mL), and borane-methyl sulfide complex (0.35 mL of 2N in
  • Step 4 Synthesis of 7-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4-dihydro-2H- benzo [b] [ 1 ,4] oxazine
  • Step 1 Synthesis of 6-bromo-7-fluoro-3-meth l-2H-benzo[b][l,4]oxazine
  • Step 3 Synthesis of 7-fluoro-3-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4- dihydro-2H-benzo[b] [ 1 ,4] oxazine
  • Aromatic bicycle (E) (LG 1 can be e.g. , CI, Br, or I; and P can be hydrogen or a protecting group) can be reacted with a boron, tin or zinc aryl or heteroaryl amine (D) (M can be -B(OR) 2 where R is hydrogen or alkyl, -Sn(alkyl) 3 , or -Zn-halo) via a palladium-mediated coupling reaction, e.g. , Suzuki, Stille, or Negishi coupling, to provide intermediate (E) with a new carbon- carbon bond formed.
  • the amino group of intermediate (E) can be reacted with a carboxylic acid, or an ester, e.g.
  • an activated ester such as an N-hydroxysuccinimide ester, or an acyl chloride, to provide compounds of Formula I.
  • 1H NMR and LCMS data for Compounds 1 to 56 is summarized below. The compounds below can be synthesized using procedures analogous to the Synthetic Examples noted in the table.
  • the Caliper LifeSciences electrophoretic mobility shift technology platform is utilized. Fluorescently labeled substrate peptide is incubated in the presence of dosed levels of compounds, a set concentration of kinase and of ATP, so that a reflective proportion of the peptide is phosphorylated. At the end of the reaction, the mix of phosphorylated (product) and non-phosphorylated (substrate) peptides are passed through the microfluidic system of the Caliper LabChip® EZ Reader II, under an applied potential difference.
  • the presence of the phosphate group on the product peptide provides a difference in mass and charge between the product peptide and the substrate peptide, resulting in a separation of the substrate and product pools in the sample. As the pools pass the LEDS within the instrument, these pools are detected and resolved as separate peaks. The ratio between these peaks therefore reflects the activity of the chemical matter at that concentration in that well, under those conditions.
  • FGFR-4 wild type assay at Km In each well of a 384-well plate, 0.5 ng/ul of wild type
  • FGFR-4 (Carna Biosciences, Inc.) was incubated in a total of 12.5 ul of buffer (100 mM HEPES pH 7.5, 0.015% Brij 35, 10 mM MgCl 2 , ImM DTT) with 1 uM CSKtide (5-FAM- KKKKEEIYFFFG-NH 2 ) and 400 uM ATP at 25 C for 90 minutes in the presence or absence of a dosed concentration series of compound (1% DMSO final concentration). The reaction was stopped by the addition of 70 ul of Stop buffer (100 mM HEPES pH 7.5, 0.015% Brij 35, 35 mM EDTA and 0.2% of Coating Reagent 3 (Caliper Lifesciences)). The plate was then read on a Caliper LabChip® EZ Reader II (protocol settings: -1.9 psi, upstream voltage -700, downstream voltage -3000, post sample sip 35s).
  • MDA-MB453 cells were plated in 96-well cell culture plates at a density of lxlO 5 cells.
  • A means that the IC 50 is less than 10 nM
  • B means the IC 50 is greater than or equal to 10 and less than ⁇
  • C means that the IC 50 is greater than or equal to 100 and less than 1000 nM
  • D means that the IC 50 is greater than 1000 nM.

Abstract

Described herein are compounds of Formula (I), or a pharmaceutically acceptable salt thereof: Formula (I) wherein: (A) denotes a single or double bond; Warhead is a moiety capable of forming a covalent bond with a nucleophile; Ring A is a 5-8 membered aryl, 5-12 membered heteroaryl, 3-7 member monocyclic or bicyclic heterocyclyl; or 3-12 membered monocyclic or bicyclic cycloalkyl group; W is C or N, X and Z are each independently CH or N; Y is CH or N-R4 where R4 is H, C1-6alkyl, or 3-12 membered cycloalkyl; each of R1-R3 is, independently, halo, cyano, optionally substituted C1-6alkyl, optionally substituted C1-6alkoxy, hydroxy, oxo, amino, amido, alkyl urea, optionally substituted 3-7 member heterocyclyl; R7 is hydrogen or C1-6alkyl; or R7 together with Ring A forms a 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R1; m is 0-5; n is 0-5; and p is 0-2 as inhibitors of FGFR-4, pharmaceutical compositions including such compounds, and methods of using such compounds and compositions to inhibit the activity of FGFR-4.

Description

HETEROBICYCLIC COMPOUNDS AND THEIR USE AS FGFR4 RECEPTOR
INHIBITORS
Claim of Priority
This application claims priority to U.S. S.N. 61/927,793, filed on January 15, 2014, which is hereby incorporated by reference in its entirety.
Field of the Invention
Described herein are compounds, pharmaceutical compositions, and methods of using such compounds and compositions to inhibit the activity of FGFR-4.
Background
Fibroblast growth factor receptor 4 (FGFR-4) is a protein that in humans is encoded by the FGFR-4 gene. This protein is a member of the fibroblast growth factor receptor family, where amino acid sequence was highly conserved between members throughout evolution. FGFR family members 1-4 differ from one another in their ligand affinities and tissue distribution. A full-length representative protein consists of an extracellular region composed of three immunoglobulin-like domains, a single hydrophobic membrane-spanning segment and a cytoplasmic tyrosine kinase domain. The extracellular portion of the protein interacts with fibroblast growth factors, setting in motion a cascade of downstream signals, ultimately influencing mitogenesis and differentiation. The genomic organization of the FGFR-4 gene encompasses 18 exons. Although alternative splicing has been observed, there is no evidence that the C-terminal half of the Iglll domain of this protein varies between three alternate forms, as indicated for FGFR 1-3.
Ectopic mineralization, characterized by inappropriate calcium-phosphorus deposition in soft tissue, has been observed in rats treated with an FGFR-1 inhibitor (Brown, AP et al. (2005), Toxicol. Pathol., p. 449-455). This suggests that selective inhibition of FGFR-4 without inhibition of other isoforms of FGFR, including FGFR-1, may be desirable in order to avoid certain toxicities. FGFR-4 preferentially binds fibroblast growth factor 19 (FGF19) and has recently been associated with the progression of certain sarcomas, renal cell cancer, breast cancer, and liver cancer. Summary of the Invention
Described herein are inhibitors of FGFR-4. Further described herein are pharmaceutical compositions and pharmaceutical formulations that include an inhibitor of FGFR-4.
In one aspect, the invention features a compound of Formula I, or pharmaceutically acceptable salt thereof, wherein:
Figure imgf000003_0001
Formula I
Figure imgf000003_0002
denotes a single or double bond;
Warhead is a moiety capable of forming a covalent bond with a nucleophile;
Ring A is a 5-8 membered aryl, 5-12 membered heteroaryl, 3-7 member heterocyclyl or 3-12 membered cycloalkyl group;
W is C or N , X and Z are each independently CH or N;
Y is CH or N-R4 wherein R4 is H, Ci_6 alkyl, or 3-12 membered cycloalkyl;
each of R 1 -R 3 is, independently, halo, cyano, optionally substituted Ci_6 alkyl, optionally substituted Ci_6 alkoxy, hydroxy, oxo, amino, amido, alkyl urea, optionally substituted 3-7 member heterocyclyl;
R 7 is hydrogen or Ci alkyl; or R 7
-6 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R1;
m is 0-5;
n is 0-5; and
p is 0-2.
In the compounds disclosed herein, a warhead is a moiety that is reactive with a nucleophile, for example, capable of forming a covalent bond with a nucleophile. Examples of warheads include, without limitation, alkyl halides, alkyl sulfonates, heteroaryl halides, epoxides, haloacetamides, maleimides, sulfonate esters, alpha-beta unsaturated ketones, alpha-beta unsaturated esters, vinyl sulfones, propargyl amides, acrylamides. In some of these instances, e.g. , acrylamide and propargyl amide, the N of the warhead is the adjacent N in the formulae shown above. Structures of exemplary warheads are shown below:
Figure imgf000004_0001
wherein X is a leaving group such as halo, or an activated hydroxyl moiety (e.g. , triflate); and each of Ra, Rb, and Rc is, independently, H, substituted or unsubstituted C1-4 alkyl, substituted or unsubstituted C1-4 cycloalkyl, or cyano.
In the formulae shown above, the warheads are typically attached to a N atom on the inhibitor. In other embodiments, the warhead can alternatively be attached to an atom other than N. Examples of exemplary warheads include, without limitation,
Figure imgf000004_0002
Figure imgf000005_0001
Other examples of warheads can be found, e.g. , in WO 2010/028236 and WO 2011/034907.
In another aspect, the invention features a method for treating a condition mediated by FGFR-4, a condition characterized by overexpression of FGFR-4, a condition characterized by amplification of FGFR4, a condition mediated by FGF19, a condition characterized by amplified FGF- 19, or a condition characterized by overexpression of FGF19, any of these methods comprising administering a therapeutically effective amount of a compound disclosed herein to a subject.
In another aspect, the invention features a method of treating any of the following conditions by administering a therapeutically effective amount of a compound disclosed herein to a subject: hepatocellular carcinoma, breast cancer, ovarian cancer, lung cancer, liver cancer, a sarcoma, or hyperlipidemia.
The invention includes all possible combinations of the embodiments described above and below.
Detailed Description
The compounds disclosed herein can form a covalent bond with FGFR-4 protein; for example, the compounds can form a covalent bond with a cysteine residue of FGFR-4, for example, the cysteine at residue 552. FGFRsl-3 do not contain this cysteine. The ability to form a covalent bond between the compound and FGFR-4 is therefore an important factor in the selectivity of the compounds disclosed herein for FGFR-4.
The details of construction and the arrangement of components set forth in the following description or illustrated in the drawings are not meant to be limiting. Other embodiments and different ways to practice the invention are expressly included. Also, the phraseology and terminology used herein are for the purpose of description and should not be regarded as limiting. The use of "including," "includes," "include," "comprising," or "having,"
"containing", "involving", and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.
Definitions
"Aliphatic group", as used herein, refers to a straight-chain, branched-chain, or cyclic hydrocarbon group and includes saturated and unsaturated groups, such as an alkyl group, an alkenyl group, and an alkynyl group.
"Alkenyl", as used herein, refers to an aliphatic group containing at least one double bond.
"Alkoxyl" or "alkoxy", as used herein, refers to an alkyl group having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
"Alkyl", as used herein, refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl- substituted cycloalkyl groups, and cycloalkyl- substituted alkyl groups. "Alkylene" refers to a double radical, that is, an aliphatic group substituted on two ends. In some embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g. , Q-C30 for straight chains, C3-C30 for branched chains), and in other embodiments can have 20 or fewer, or 10 or fewer. Likewise, certain cycloalkyls may have from 3- 10 carbon atoms in their ring structure, and in some embodiments may have 5, 6 or 7 carbons in the ring structure. The term "alkenyl", as used herein, refers to an aliphatic group containing at least one double bond; the term "alkynyl", as used herein, refers to an aliphatic group containing at least one triple bond.
"Alkylthio", as used herein, refers to a hydrocarbyl group having a sulfur radical attached thereto. In some embodiments, the "alkylthio" moiety is represented by one of -S-alkyl, -S- alkenyl, or -S-alkynyl. Representative alkylthio groups include methylthio, ethylthio, and the like.
"Amido", as used herein, refers to -C(=0)-N(R1)( R2) or -N(R1)-C(=0)-R2 wherein each
1 2
of R and R is independently hydrogen, hydroxyl, C C6 alkyl, or C C6 alkoxyl, e.g. , methoxyl.
"Amino", as used herein, refers to -NH2, -NH(alkyl), or -N(alkyl) (alkyl). "Amplified", as used herein, means additional copies of a gene or chromosome segment are produced in cancer cells that may confer a growth or survival advantage.
"Aralkyl", as used herein, refers to an alkyl group substituted with an aryl group (e.g. , an aromatic or heteroaromatic group).
"Aryl", as used herein, refers to 5-, 6-, and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, phenyl, pyrrolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyrazolyl, pyridinyl, pyrazinyl, pyridazinyl and pyrimidinyl, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles" or "heteroaromatics." The aromatic ring can be substituted at one or more ring positions with such substituents as described above, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, polycyclyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or
heteroaromatic moieties, -CF3, -CN, or the like. The term "aryl" also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are "fused rings") wherein at least one of the rings is aromatic, e.g. , the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls. Each ring can contain, e.g. , 5-7 members.
The term "carbocycle" as used herein, refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon.
The term "cycloalkyl" as used herein, includes cyclic, bicyclic, tricyclic, or polycyclic non-aromatic hydrocarbon groups having 3 to 12 carbons. Any substitutable ring atom can be substituted (e.g. , by one or more substituents). The cycloalkyl groups can contain fused or spiro rings. Fused rings are rings that share a common carbon atom. Examples of cycloalkyl moieties include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl,
methylcyclohexyl, adamantyl, and norbornyl.
"Covalent inhibitor," as used herein, means an inhibitor that can form a covalent bond with a protein.
The "enantiomeric excess" or "% enantiomeric excess" of a composition can be calculated using the equation shown below. In the example shown below a composition contains 90% of one enantiomer, e.g., the S-enantiomer, and 10% of the other enantiomer, i.e., the R- enantiomer.
ee = (90-10)/100 = 80%.
Thus, a composition containing 90% of one enantiomer and 10% of the other enantiomer is said to have an enantiomeric excess of 80%. In some embodiments, the compositions described herein contain an enantiomeric excess of at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the S-enantiomer. In other words, the compositions contain an enantiomeric excess of the S-enantiomer over the R-enantiomer. In some
embodiments, the compositions described herein contain an enantiomeric excess of at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the R- enantiomer. In other words, the compositions contain an enantiomeric excess of the R- enantiomer over the S-enantiomer.
"FGFR-4" or "FGFR-4 protein" refers to any form of the FGFR-4 protein, including wild type and all variant forms (including, without limitation, mutant forms and splice variants). The FGFR-4 protein is a product of the FGFR-4 gene, and the FGFR-4 protein therefore includes any protein encoded by any form of the FGFR-4 gene, including all aberrations, e.g., point mutations, indels, translocation fusions, and focal amplifications.
"Halo" refers to a radical of any halogen, e.g., -F, -CI, -Br, or -I.
"Haloalkyl" and "haloalkoxy" refers to alkyl and alkoxy structures that are substituted with one or more halo groups or with combinations thereof. For example, the terms
"fluoroalkyl" and "fluoro alkoxy" include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine.
"Heteroarylalkyl" refers to an alkyl group substituted with a heteroaryl group.
"Heterocyclyl" or "heterocyclic group" refers to a ring structure, such as a 3- to 14- membered ring structure, whose ring(s) include one or more heteroatoms. Heterocyclyl can also include polycycles, with each group having, e.g., 3-7 ring members. The term "heterocyclyl" or "heterocyclic group" includes "heteroaryl" and "saturated or partially saturated heterocyclyl" structures. The term "saturated or partially saturated heterocyclyl" refers to a non-aromatic cyclic structure that includes at least one heteroatom. The term "heterocyclyl" can also include 8-12 membered bicyclic heterocyclyls, e.g., wherein a saturated or partially saturated
heterocyclyl is fused to an aromatic or heteroaromatic ring. The term "heterocyclyl" can also include 8- 12 membered bicyclic heterocyclyls, e.g. , wherein a saturated or partially saturated cycloalkyl is fused to an aromatic or heteroaromatic ring. The point of attachment of the heterocyclyl to the rest of the molecule can be through the saturated or partially saturated heterocyclyl or cycloalkyl, or through the aromatic or heteroaromatic ring.
"Heteroaryl" refers to a 5-14 membered (i.e. , 5-8 membered monocyclic, 8- 12 membered bicyclic, or 11-14 membered tricyclic) aromatic ring system having 1-3 ring heteroatoms if monocyclic, 1-6 ring heteroatoms if bicyclic, or 1-9 ring heteroatoms if tricyclic, said ring heteroatoms independently selected from O, N, and S (e.g. , 1-3, 1-6, or 1-9 ring heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively). Any ring atom can be substituted (e.g. , by one or more substituents).
Heterocyclyl groups include, for example, thiophenyl, thianthrenyl, furanyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathiin, pyrrolyl, imidazolyl, pyrazolyl, isothiazolyl, isoxazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, quinolizinyl, isoquinolinyl, quinolinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, quinazolinyl, cinnolinyl, pteridinyl, carbazolyl, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, oxolane, thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. The heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, - CN, or the like.
"Heterocyclylalkyl" refers to an alkyl group substituted with a heterocycle group.
"Hydroxy" or "hydroxyl" refers to the chemical group -OH.
"Inhibitor" refers to a compound that inhibits an enzyme such that a reduction in activity of the enzyme can be observed, e.g. , in a biochemical assay. In certain embodiments, an inhibitor has an IC50 of less than about 1 μΜ, less than about 500 nM, less than about 250 nM, less than about 100 nM, less than about 50 nM, or less than about 10 nM. An inhibitor of FGFR- 4 refers to a compound that inhibits FGFR-4. "Nucleophile" refers to a species that donates an electron-pair to an electrophile to form a chemical bond in a reaction. In some embodiments, a nucleophile can be an oxygen nucleophile, e.g. , water or hydroxyl, a nitrogen nucleophile, e.g. , amine, or a sulfur nucleophile, e.g. , thiol, such as, for example, the thiol in the side chain of a cysteine residue.
"Overexpressed," as used herein, means there is production of a gene product in a sample that is substantially higher than that observed in a population of control samples (e.g. normal tissue).
"Oxo" refers to (=0).
"Selective" refers to a compound that inhibits the activity of a target protein, e.g. , FGFR- 4, more potently than it inhibits activity of other proteins. In this instance, the isoforms FGFR- 1, FGFR-2, FGFR-3, and FGFR-4 are all considered distinct proteins. In some embodiments, a compound can inhibit the activity of the target protein, e.g. , FGFR-4, at least 1.5, at least 2, at least 5, at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 200, at least 500, or at least 1000 or more times potently than it inhibits the activity of a non-target protein.
"Substituted" refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that "substitution" or "substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g. , which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. For instance, the substituents of a substituted alkyl may include substituted and unsubstituted forms of amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), -CF3, -CN and the like. Exemplary substituted alkyls are described below. Cycloalkyls can be further substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, -CF3, -CN, and the like. Analogous substitutions can be made to alkenyl and alkynyl groups to produce, for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls, iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls.
As used herein, the definition of each expression, e.g. , alkyl, m, n, etc., when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
"Warhead moiety" or "warhead" refers to a moiety of an inhibitor which participates, either reversibly or irreversibly, with the reaction of a donor, e.g. , a protein, with a substrate. Warheads may, for example, form covalent bonds with the protein, or may create stable transition states, or be a reversible or an irreversible alkylating agent. For example, the warhead moiety can be a functional group on an inhibitor that can participate in a bond-forming reaction, wherein a new covalent bond is formed between a portion of the warhead and a donor, for example an amino acid residue of a protein. In embodiments, the warhead is an electrophile and the "donor" is a nucleophile such as the side chain of a cysteine residue. Examples of suitable warheads include, without limitation, the groups shown below:
Figure imgf000011_0001
wherein X is a leaving group such as halo, or an activated hydroxyl moiety (e.g. , triflate); and each of Ra, Rb, and Rc is, independently, H, substituted or unsubstituted C1-4 alkyl, substituted or unsubstituted Ci_4 cycloalkyl, or cyano.
As used herein, the term "patient" or "subject" refers to organisms to be treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g. , murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans.
As used herein, the term "effective amount" refers to the amount of a compound (e.g. , a compound of the present invention) sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. As used herein, the term "treating" includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
As used herein, the term "modulate" or "modulating" refers to an increase or decrease, e.g. , in the activity of an enzyme in response to exposure to a compound or composition described herein, e.g. , the inhibition of FGFR-4, in at least a sub-population of cells in a subject such that a desired end result is achieved (e.g. , a therapeutic result). In some embodiments, a compound as described herein inhibits a target described herein, e.g. , FGFR-4.
Compounds
The compounds described herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H) or carbon- 14 (14C). All isotopic variations of the compounds disclosed herein, whether radioactive or not, are intended to be encompassed within the scope of the present invention. For example, deuterated compounds
13
or compounds containing C are intended to be encompassed within the scope of the invention.
Certain compounds can exist in different tautomeric forms, and all possible tautomeric forms of all of the compounds described herein are intended to be encompassed within the scope of the invention. Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g. , enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The compounds described herein can be useful as the free base or as a salt.
Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like. (See, for example, Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66: 1- 19.)
Certain compounds disclosed herein can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention. Certain compounds disclosed herein may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
In one aspect, the invention provides a compound having structural Formula I:
Figure imgf000013_0001
Formula I
wherei
Figure imgf000013_0002
denotes a single or double bond; Warhead is a moiety capable of forming a covalent bond with a nucleophile;
Ring A is a 5-8 membered aryl, 5-12 membered heteroaryl, 3-7 member heterocyclyl or 3-12 membered cycloalkyl group;
W is C or N, X and Z are each independently CH or N;
Y is CH or N-R4 where R4 is H, C1-6 alkyl, or 3-12 membered cycloalkyl;
each of R 1 -R 3 is, independently, halo, cyano, optionally substituted C1-6 alkyl, optionally substituted Ci_6 alkoxy, hydroxy, oxo, amino, amido, alkyl urea, optionally substituted 3-7 member heterocyclyl;
R 7 is hydrogen or C1-6 alkyl; or R 7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R1;
m is 0-5;
n is 0-5; and
p is 0-2.
In some embodiments, denotes a In some embodiments, denotes a double bond. In som
Figure imgf000014_0001
e embodiments, gle bond and the other is a double bond.
In some embodiments, Ring A is aryl. In some embodiments, Ring A is phenyl substituted with 0-3 R1. In some embodiments, R1 is selected from the group consisting of halo, optionally substituted C1-6 alkyl, and optionally substituted C1-6 alkoxy.
In some embodiments, R 7 is hydrogen or Ci_6 alkyl. In some embodiments, R 7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R 11. In some embodiments, R 7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-3 occurrences of R1.
In some embodiments, X is N. In some embodiments, X is CH.
In some embodiments, Y is N-R4 wherein R4 is Ci_6 alkyl or 3-12 membered cycloalkyl. In some embodiments, R4 is methyl, ethyl or isopropyl. In some embodiments, R4 is ethyl. In some embodiments, R4 is 3-12 membered cycloalkyl. In some embodiments, R4 is cyclopentyl.
In some embodiments, Y is N-R4 wherein R4 is Ci_6 alkyl or 3-12 membered cycloalkyl and R3 is oxo. In some embodiments, R4 is methyl, ethyl or isopropyl. In some embodiments, R4 is ethyl. In some embodiments, R4 is 3-12 membered cycloalkyl. In some embodiments, R4 is cyclopentyl.
In some embodiments, q is 0.
In some embodiments, the warhead moiety is selected from the group consisting of
Figure imgf000015_0001
wherein each of Ra, R , and Rc is, independently, hydrogen, optionally substituted C1-4 alkyl, optionally substituted 3-12 membered cycloalkyl, or cyano.
In some embodiments, R 2 is alkoxy or halo. In some embodiments, R 2 is methoxy or chloro. In some embodiments, two R 2 are chloro and two R 2 are methoxy.
In one embodiment, the invention features a compound of Formula 1(a) or a
pharmaceutically acceptable salt thereof:
Figure imgf000015_0002
Formula 1(a)
wherein X is CH or N. In some embodiments, X is CH. In some embodiments, X is
In one embodiment, the invention features a compound of Formula 1(b) or a
pharmaceutically acceptable salt thereof:
Figure imgf000016_0001
Formula 1(b)
wherein X is CH or N. In some embodiments, X is CH. In some embodiments, X is
In one embodiment, the invention features a compound of Formula 1(b) or a pharmaceutically acceptable salt thereof:
Figure imgf000016_0002
Formula 1(c)
wherein X is CH or N. In some embodiments, X is CH. In some embodiments, X is
In one embodiment, the invention features a compound of Formula 1(d) or a pharmaceutically acce table salt thereof:
Figure imgf000016_0003
Formula 1(d). In some embodiments, the Warhead moiety is
Figure imgf000017_0001
Ring A is phenyl, and R7 together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R ; R is C1-6 alkyl or 3-12 membered cycloalkyl; R is Ci_6 alkoxy or halo; and n is 4. In some embodiments, R together with Ring A forms an 8-12 membered bicyclic heterocyclyl optionally substituted with 1-3 occurrences of R1.
In some embodiments, Ring A together with R forms an 8-10 membered heterocyclyl. In some embodiments, Ring A together with R forms:
Warhead
Figure imgf000017_0002
wherein V is CH2 or O; r is 0 or 1; and R1 and m are as defined herein.
7
In some embodiments, Ring A together with R forms:
ng A
s
Figure imgf000017_0003
forms: . In some embodiments, Ring A together with R7 forms:
Figure imgf000018_0001
. In some embodiments,
Figure imgf000018_0002
Ring A together with R forms:
wherein V is CH2 or O; r is 0 or 1 ; and R and m are as defined herein.
The invention also features pharmaceutical compositions comprising a pharmaceutically acceptable carrier and any compound of Formulas 1, 1(a), 1(b), 1(c), and 1(d).
Pharmaceutical Compositions
While it is possible for a compound disclosed herein to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation, where the compound is combined with one or more pharmaceutically acceptable excipients or carriers. The compounds disclosed herein may be formulated for administration in any convenient way for use in human or veterinary medicine. In certain embodiments, the compound included in the pharmaceutical preparation may be active itself, or may be a prodrug, e.g. , capable of being converted to an active compound in a physiological setting. In certain embodiments, the compounds provided herein include their hydrates.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
Examples of pharmaceutically acceptable salts of a compound described herein include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
Examples of suitable acid salts include acetate, adipate, benzoate, benzenesulfonate, butyrate, citrate, digluconate, dodecylsulfate, formate, fumarate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactate, maleate, malonate,
methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, palmoate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, tosylate and undecanoate. Salts derived from appropriate bases include alkali metal (e.g. , sodium), alkaline earth metal (e.g. , magnesium), ammonium and N-(alkyl)4 + salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds described herein. Water or oil-soluble or dispersible products may be obtained by such quaternization.
Examples of pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; (21) cyclodextrins such as Captisol®; targeting ligands attached to nanoparticles, such as Accurins™; and (22) other non-toxic compatible substances, such as polymer-based compositions, employed in pharmaceutical formulations.
Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium
metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid,
ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Solid dosage forms (e.g. , capsules, tablets, pills, dragees, powders, granules and the like) can include one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. Liquid dosage forms can include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect.
Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required.
When the compounds disclosed herein are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
The formulations can be administered topically, orally, transdermally, rectally, vaginally, parentally, intranasally, intrapulmonary, intraocularly, intravenously, intramuscularly, intraarterially, intrathecally, intracapsularly, intradermally, intraperitoneally, subcutaneously, subcuticularly, or by inhalation.
Indications
FGFR-4 regulates proliferation, survival, and alpha-fetoprotein secretion during hepatocellular carcinoma (HCC) progression; inhibitors of FGFR-4 are therefore promising potential therapeutic agents for this unmet medical need (Ho et al., Journal of Hepatology, 2009, 50: 118-27). HCC afflicts more than 550,000 people worldwide every year and has one of the worst 1-year survival rates of any cancer type.
Further evidence of the link between FGFR-4 and HCC is shown through the
involvement of FGF 19, a member of the fibroblast growth factor (FGF) family, which consists of hormones that regulate glucose, lipid, and energy homeostasis. Increased hepatocyte
proliferation and liver tumor formation have been observed in FGF 19 transgenic mice. FGF 19 activates FGFR-4, its predominant receptor in the liver, and it is believed that activation of FGFR-4 is the mechanism whereby FGF 19 can increase hepatocyte proliferation and induce hepatocellular carcinoma formation (Wu et al., J Biol Chem (2010) 285(8):5165-5170). FGF 19 has been identified as a driver gene in HCC by others as well (Sawey et al., Cancer Cell (2011) 19: 347-358). It is therefore believed that the compounds disclosed herein, which are potent and selective inhibitors of FGFR-4, can be used to treat HCC and other liver cancers.
Oncogenome screening has identified an activating fibroblast growth factor receptor 4 (FGFR-4) Y367C mutation in the human breast cancer cell line MDA-MB-453. This mutation was shown to elicit constitutive phosphorylation, leading to an activation of the mitogen- activated protein kinase cascade. Accordingly, it has been suggested that FGFR-4 may be a driver of tumor growth in breast cancer (Roidl et al., Oncogene (2010) 29(10): 1543-1552). It is therefore believed that the compounds disclosed herein, which are potent and selective inhibitors of FGFR-4, can be used to treat FGFR-4 modulated breast cancer.
Molecular changes (e.g. , translocations) in genes upstream of FGFR-4 can lead to activation/overexpression of FGFR-4. For example, a PAX3-FKHR translocation/gene fusion can lead to FGFR-4 overexpression. Overexpression of FGFR-4 due to this mechanism has been associated with rhabdomyosarcoma (RMS) (Cao et al., Cancer Res (2010) 70(16): 6497-6508).
Mutations in FGFR-4 itself (e.g. , kinase domain mutations) can lead to over-activation of the protein; this mechanism has been associated with a subpopulation of RMS (Taylor et al., J Clin
Invest (2009) 119: 3395-3407). It is therefore believed that the compounds disclosed herein, which are potent and selective inhibitors of FGFR-4, can be used to treat FGFR-4 modulated
RMS and other sarcomas.
Other diseases have been associated with changes in genes upstream of FGFR-4 or with mutations in FGFR-4 itself. For example, mutations in the kinase domain of FGFR-4 lead to over-activation, which has been associated with lung adenocarcinoma (Ding et al., Nature (2008) 455(7216): 1069- 1075). Amplification of FGFR-4 has been associated with conditions such as renal cell carcinoma (TCGA provisional data). In addition, silencing FGFR4 and inhibiting ligand-receptor binding significantly decrease ovarian tumor growth, suggesting that inhibitors of FGFR4 could be useful in treating ovarian cancer. (Zaid et al., Clin. Cancer Res. (2013) 809).
Pathogenic elevations of bile acid levels have been linked to variations in FGF19 levels (Vergnes et al., Cell Metabolism (2013) 17, 916-28). Reduction in the level of FGF19 may therefore be of benefit in promoting the synthesis of bile acid and thus in the treatment of hyperlipidemia.
Dose Levels
Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
The selected dosage level will depend upon a variety of factors including the activity of the particular compound disclosed herein employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
In general, a suitable daily dose of a compound of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
Combination and Targeted Therapy
Administration of the FGFR-4 inhibitors disclosed herein can be combined with other cancer treatments. For example, the inhibitors can be administered in combination with surgical treatments, radiation, or other therapeutic agents such as antibodies, other selective kinase inhibitors, or chemotherapeutics. The inhibitors may also be administered in combination with RNAi therapy or antisense therapy. The FGFR-4 inhibitors described herein may be combined with one, two, or more other therapeutic agents. Examples of other therapeutic agents include sorafenib, gemcitabine, capecitabine, and doxorubicin. In the examples outlined below, it is understood that "second therapeutic agent" also includes more than one therapeutic agent other than the FGFR-4 inhibitor. A FGFR-4 inhibitor described herein may be administered with one, two, or more other therapeutic agents.
The FGFR-4 inhibitors described herein and the second therapeutic agent do not have to be administered in the same pharmaceutical composition, and may, because of different physical and chemical characteristics, be administered by different routes. For example, the FGFR-4 inhibitor can be administered orally, while the second therapeutic agent is administered intravenously. The determination of the mode of administration and the advisability of administration, where possible, in the same pharmaceutical composition, is well within the knowledge of the skilled clinician. The initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician.
The FGFR-4 inhibitor and the second therapeutic agent may be administered
concurrently (e.g. , simultaneously, essentially simultaneously or within the same treatment protocol) or sequentially (i.e. , one followed by the other, with an optional time interval in between), depending upon the nature of the proliferative disease, the condition of the patient, and the actual choice of second therapeutic agent to be administered.
In addition, the FGFR-4 inhibitors disclosed herein can be administered as part of an antibody-drug conjugate, where the FGFR-4 inhibitor is the "payload" portion of the conjugate. The table below shows exemplary compounds.
Figure imgf000024_0001
Figure imgf000025_0001

Figure imgf000026_0001

Figure imgf000027_0001

Figure imgf000028_0001

Figure imgf000029_0001

Figure imgf000030_0001

Figure imgf000031_0001
30
Figure imgf000032_0001
31
Figure imgf000033_0001
32
Figure imgf000034_0001
33
Figure imgf000035_0001
Figure imgf000036_0001

Figure imgf000037_0001

Figure imgf000038_0001

Figure imgf000039_0001

Figure imgf000040_0001
Figure imgf000041_0001
40
Figure imgf000042_0001
Synthesis
Compounds of the invention, including salts and N -oxides thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes, such as those in the Schemes below. The reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g. , temperatures which can range from the solvent' s freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.
Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4th ed., John Wiley & Sons: New Jersey, (2006), which is incorporated herein by reference in its entirety.
Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic
1 13
resonance (NMR) spectroscopy (e.g. , H or C), infrared (IR) spectroscopy, spectrophotometry (e.g. , UV- visible), mass spectrometry (MS), or by chromatographic methods such as high performance liquid chromatography (HPLC) or thin layer chromatography (TLC).
LCMS: Liquid chromatography-mass spectrometry (LCMS) data (sample analyzed for purity and identity) can be obtained with an Agilent model- 1260 LC system using an Agilent model 6120 mass spectrometer utilizing ES-API ionization fitted with an Agilent Poroshel 120 (EC-C18, 2.7um particle size, 3.0 x 50mm dimensions) reverse-phase column at 22.4 degrees Celsius. The mobile phase can be of a mixture of solvent 0.1% formic acid in water and 0.1% formic acid in acetonitrile. A constant gradient from 95% aqueous/5% organic to 5%
aqueous/95% organic mobile phase over the course of 4 minutes can be utilized.
Proton NMR: 1H NMR spectra can be obtained with a Varian 400MHz Unity Inova 400 MHz NMR instrument (acquisition time = 3.5 seconds with a 1 second delay; 16 to 64 scans). Where characterized, all protons are generally reported in DMSO-d6 solvent as parts-per million (ppm) with respect to residual DMSO (2.50 ppm).
The below Schemes are meant to provide general guidance in connection with preparing the compounds of the invention. One skilled in the art would understand that the preparations shown in the Schemes can be modified or optimized using general knowledge of organic chemistry to prepare various compounds of the invention.
Synthetic Example 1: Synthesis of 2-chloro-6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazoline Step 1: Synthesis of (2-amino-5-bromo henyl)methanol
Figure imgf000044_0001
To a solution of 2-amino-5-bromobenzoic acid (10.0 g, 46.3 mmol) in THF (150 mL) was added BH3-THF (1 M, 231 mL) at room temperature, and the reaction mixture was stirred overnight. An aliquot of the reaction mixture was analyzed by LCMS and indicated that the reaction had proceeded to completion. The reaction was quenched with water (150 mL) and extracted with EtOAc (3 x 500 mL). The organic layers were separated, combined, washed with water (200 mL) and brine (200 mL), dried over sodium sulfate, filtered, and concentrated to afford the title compound (10 g, crude), which was directly used in the next step without further purification. MS (ES+) C7H8BrNO requires: 201, found: 202, 204 [M + H]+.
Step 2: Synthesis of 2-amino-5-bromobenzaldehyde
Figure imgf000044_0002
A mixture of (2-amino-5-bromophenyl)methanol (10 g, 49.5 mmol) and Mn02 (25.8 g, 296.6 mmol) in CH2C12 (400 mL) was stirred at RT overnight. LCMS showed the reaction was completed. The solid was filtered off, and the filtrate was concentrated to give the title compound as a light yellow solid (8 g, 81%), which was directly used in next step without further purification. MS (ES+) C7H6BrNO requires: 199, found: 200, 202 [M + H]+.
Step 3: Synthesis of 6-bromo uinazolin-2-ol
Figure imgf000044_0003
A mixture of 2-amino-5-bromobenzaldehyde (29) (6 g, 30.0 mmol) and urea (30) (27 g, 450.0 mmol) was heated to 180 °C and stirred for 5 hours. LCMS showed the reaction was completed. The reaction mixture was cooled to room temperature, and the resulting precipitate was washed with H20 (3 x 500 mL) and co-evaporated with toluene three times to completely remove the moisture trapped. 6-bromoquinazolin-2-ol (31)(6 g, 89%) was obtained as a yellow solid. MS (ES+) C8H5BrN20 requires: 224, found: 225, 227 [M + H]+.
Step 4: Synthesis of 6-bromo-2-chloro uinazoline
Figure imgf000045_0001
A solution of 6-bromoquinazolin-2-ol (31) (6.0 g, 26.7 mmol) in POCI3 (80 mL) was refluxed at 110 °C for 5 hours. An aliquot of the reaction mixture was analyzed by LCMS and indicated that the reaction had proceeded to completion. Most of POCI3 was removed under reduced pressure, and the residue was added dropwise to ice water (500 mL). The resulting precipitate was collected via filtration as a yellow solid (3.5 g, 54%). MS (ES+) C8H4BrClN2 requires: 242, found: 243, 245 [M + H]+.
Step 5: S nthesis of 2-chloro-6-(3,5-dimethox henyl)quinazoline
Figure imgf000045_0002
A mixture of 6-bromo-2-chloroquinazoline (32) (5.0 g, 20.5 mmol), 3,5- dimethoxyphenylboronic acid (33) (3.7 g, 20.5 mmol), CS2CO3 (20.0 g, 61.5 mmol) and
Pd(PPh3)2Cl2 (1.4 g, 2.1 mmol) in THF (50 mL), dioxane (50 mL) and water (10 mL) was degassed with N2 three times, and stirred at 80 °C for 3 hours. An aliquot of the reaction mixture was analyzed by both TLC and LCMS, which indicated that the reaction had proceeded to completion. The mixture was cooled to room temperature, and extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with water and brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 8: 1) to obtain 2-chloro-6-(3,5-dimethoxyphenyl)quinazoline (34) as a light yellow solid (2.4 g, 38%). MS (ES+) Ci6H13ClN202 requires: 300, found: 301, 303 [M + H]+. Step 6: Synthesis of 2-chloro-6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazoline
Figure imgf000046_0001
To a solution of 2-chloro-6-(3,5-dimethoxyphenyl)quinazoline (34) (2.7 g, 8.9 mmol) in dry THF (80 mL) was added dropwise SO2CI2 (3.0 g, 22.3 mmol) at -20 °C, and the reaction mixture was stirred for an additional hour. An aliquot of the reaction mixture was analyzed by both TLC and LCMS, which indicated that the reaction had proceeded to completion. The reaction mixture was quenched with water (1 mL), and the solvents were removed under reduced pressure. The precipitate was washed with CH3CN and dried to obtain 2-chloro-6-(2,6-dichloro- 3,5-dimethoxyphenyl)quinazoline (35) (2.6 g, 79%) as a white solid.
Synthetic Example 2: Synthesis of 7-chloro-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl- L6-naphthyridin-2(lH)-one
Step 1: Synthesis of eth l 6-chloro-4-(methylamino)nicotinate
Figure imgf000046_0002
overnight H
To a solution of ethyl 4,6-dichloronicotinate (5.0 g, 22.7 mmol) in acetonitrile (50 mL) was added methylamine hydrochloride salt (1.84 g, 27.2 mmol) and diisopropylethylamine (14.6 g, 113.6 mmol), and the reaction mixture was heated at 70 °C overnight. LCMS showed the reaction was completed. The reaction was cooled to RT, quenched with water (50 mL) and extracted with ethyl acetate (3 x 100 mL). The organic layers were separated, combined, washed with water (50 mL) and brine (100 mL), dried over sodium sulfate, filtered, and concentrated to afford the title compound (4.7 g, crude), which was directly used in the next step without further purification. MS (ES+) C9H11CIN2O2 requires: 214, 216, found: 215, 217 [M + H]+.
Step 2: Synthesis of 6-chloro-4-(methylamino)pyridin-3-yl)methanol
Figure imgf000046_0003
To a solution of ethyl 6-chloro-4-(methylamino)nicotinate (4.7 g, 21.9 mmol) in THF (30 mL) and methanol (30 mL) was added lithium borohydride (2.4 g, 109.8 mmol), and the reaction mixture was heated at 55 °C overnight. LCMS showed the reaction was completed. The reaction was cooled to RT, quenched with water (1 mL) and filtered. The filtrate was concentrated to afford the title compound (4.2 g, crude) as a white solid, which was directly used in the next step without further purification. MS (ES+) C7H9C1N20 requires: 172, 174, found: 173, 175 [M + H]+.
Step 3: Synthesis o -chloro-4-(methylamino)nicotinaldehyd
Figure imgf000047_0001
A mixture of (6-chloro-4-(methylamino)pyridin-3-yl)methanol (4.2 g, 24.7 mmol) and manganese(IV) oxide (active, 25.8 g, 296.6 mmol) in dichloromethane (50 mL) and THF (50 mL) was stirred at RT overnight. LCMS showed the reaction was completed. The solid was filtered off, and the filtrate was concentrated to afford the title compound (3.7 g, crude) as a light yellow solid, which was directly used in the next step without further purification. MS (ES+) C7H7C1N20 requires: 170, 172, found: 171, 173 [M + H]+.
Step 4: Synthesis of 7-chloro-3-(3,5-dimethoxyphenyl)-l-methyl-l,6-naphthyridin-2(lH)-one
Figure imgf000047_0002
A mixture of 6-chloro-4-(methylamino)nicotinaldehyde (3.7 g, 21.7 mmol), methyl 2-(3,5- dimethoxyphenyl) acetate (4.5 g, 21.7 mmol) and potassium carbonate (9.0 g, 65.1 mmol) in N,N- dimethylformamide (30 mL) was heated at 105 °C for 5 h. LCMS showed the reaction was completed. The reaction was cooled to RT, quenched with water (200 mL), and filtered. The filtration cake was washed by petroleum ether (50 mL) and ethyl acetate (50 mL) to afford the title compound (5.8 g, 77%) as a yellow solid. MS (ES+) C18H19C1N203 requires: 346, 348, found: 347, 349 [M + H]+.
Step 5: Synthesis of 7-chloro-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl-l,6-naphthyridin- 2(lH)-one
Figure imgf000048_0001
To a solution of 7-chloro-3-(3,5-dimethoxyphenyl)-l-methyl-l,6-naphthyridin-2(lH)-one (5.6 g, 16.9 mmol) in actonitrile (30 mL) was dropwise added sulfuryl chloride (3.36 mL, 42.2 mmol) at -20 °C, and the mixture was stirred for another 15 mins. LCMS showed the reaction was completed. The reaction was quenched with water (1 mL), and the solvents were removed under reduced pressure. The precipitate was washed with acetonitrile and dried to afford the title compound (5.01 g, 75%) as a white solid.
Synthetic Example 2a: Synthesis of 7-chloro-l-isopropyl-3-(2-fluoro-3,5-dimethoxyphenyl)-l,6- naphthyridin-2(lH)-one
Figure imgf000048_0002
To a stirred -15°C mixture of 7-chloro-3-(3,5-dimethoxyphenyl)-l-isopropyl-l,6- naphthyridin-2(lH)-one (205.5 mg, 0.573 mmol) in acetonitrile (11.5 mL) was added Selectfluor (203 mg, 0.573 mmol). The mixture was stirred at -15°C and slowly allowed to warm up to room temperature over 1 hour. The reaction was stirred for a total of 3 hours and 45 minutes before workup. The reaction was diluted with dichloromethane (50 mL) and washed with saturated aqueous NaHC03 (30 mL). The aqueous layer was extracted with fresh
dichloromethane (2 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated down and purified by silica gel chromatography to yield 7-chloro-l- isopropyl-3-(2-fluoro-3,5-dimethoxyphenyl)-l,6-naphthyridin-2(lH)-one (124.9 mg, 57.3% yield). MS: M+l = 377. Synthetic Example 2b: Synthesis of 7-chloro-3-(2-chloro-6-fluoro-3,5-dimethoxyphenyl)-l- ethyl- 1 ,6-naphthyridin-2( lH)-one
Step 1: Synthesis of 7-chloro-l-ethyl-3-(2-fluoro-3,5-dimethoxyphenyl)-l,6-naphthyridin-2(lH)- one
Figure imgf000049_0001
7-Chloro- l-ethyl-3-(2-fluoro-3,5-dimethoxyphenyl)- 1 ,6-naphthyridin-2( lH)-one was prepared a similar manner as 7-chloro-l-isopropyl-3-(2-fluoro-3,5-dimethoxyphenyl)-l,6-naphthyridin- 2(lH)-one from 7-chloro-3-(3,5-dimethoxyphenyl)-l-ethyl-l,6-naphthyridin-2(lH)-one in Synthetic Example 2a. MS: M+l = 363.
Step 2: Synthesis of 7-chloro-3-(2-chloro-6-fluoro-3,5-dimethoxyphenyl)-l-ethyl-l,6- naphthyridin-2(lH)-one
Figure imgf000049_0002
To a 0°C stirred suspension of 7-chloro-l-ethyl-3-(2-fluoro-3,5-dimethoxyphenyl)-l,6- naphthyridin-2(lH)-one (164.6 mg, 0.422 mmol) in acetonitrile (7 mL) was added sulfuryl chloride (57 mg, 0.422 mmol). The mixture was stirred at 0°C. At about 5 minutes, the reaction was quenched by addition of water (-30 mL). After 10 minutes of stirring, the yellow
suspension was extracted with dichloromethane (120 mL + 60 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated down, and dried. The crude yellow thick oil was purified by silica gel chromtaography to yield 7-chloro-3-(2-chloro-6-fluoro-3,5- dimethoxyphenyl)-l-ethyl-l,6-naphthyridin-2(lH)-one (88 mg, 50% yield) as a yellow solid. MS: M+l = 397. Synthetic Example 3: Synthesis of 7-chloro-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-ethyl-3,4- dihydropyridor4,3-dlpyrimidin-2(lH -one
Figure imgf000050_0001
Figure imgf000050_0002
Step 1: Synthesis of ethyl 6-chloro-4-(ethylamino)nicotinate
Figure imgf000050_0003
To a solution of ethyl 4,6-dichloronicotinate (6.0 g, 27.5 mmol) in acetonitrile (120 mL) was added ethylamine hydrochloride (1.45 g, 33.0 mmol) and DIPEA (17.7 g, 137.5 mmol) at RT. The resultant reaction mixture was heated at 70 °C for 12 h. After that, the reaction mixture was quenched with water (50 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL). The organic layers were separated, combined, dried over sodium sulfate, filtered, and
concentrated to afford 6-chloro-4-(ethylamino)nicotinate (6.0 g, crude), which was directly used in next step without further purification. MS (ES+) CioHnClNiOi requires: 228, 230, found: 229, 231 [M + H]+. Step 2: Synthesis of (6-chloro-4-(ethylamino)pyridin-3-yl)methanol
Figure imgf000050_0004
To a solution of ethyl 6-chloro-4-(ethylamino)nicotinate (3.0 g, 13.2 mmol) in THF (30 mL) and MeOH (30 mL) was added lithium borohydride (1.4 g, 65.8 mmol) at RT. The resultant reaction mixture was heated at 55 °C for 12 h. After that, the reaction mixture was quenched with water (1 mL), and the precipitate was filtered off. The filtrate was concentrated to give title compound 6-chloro-4-(ethylamino)nicotinaldehyde (2.1 g, crude), which was directly used in next step without further purification. MS (ES+) C8HnClN20 requires: 186, 188, found: 187, 189 [M + H]+.
Step 3: Synthesis of 6-chloro-4-(ethylamino)nicotinaldehyde (Notebook: SP-0010571-035):
Figure imgf000051_0001
A mixture of (6-chloro-4-(ethylamino)pyridin-3-yl)methanol (4.4 g, 23.7 mmol) and manganese(IV) oxide (20.5 g, 236.6 mmol) in DCM (250 mL) and THF (50 mL) was stirred at RT for 12 h. LCMS showed the reaction was completed. The solid was filtered off, and the filtrate was concentrated to give 6-chloro-4-(ethylamino)nicotinaldehyde (4.0 g) as a light yellow solid, which was directly used in next step without the further purification. MS (ES+)
C8H9C1N20 requires: 184, 186, found: 187, 189 [M + H]+.
Step 4: Synthesis of ethyl 2-chloro-5-((3,5-dimethoxyphenylamino)methyl)-N-ethylpyridin-4- amine
Figure imgf000051_0002
A mixture of 6-chloro-4-(ethylamino)nicotinaldehyde (700 mg, 3.8 mmol), 3,5- dimethoxyaniline (872 mg, 5.7 mmol) in MeOH (10 mL) was stirred at 20 °C for 4 h. After that, sodium cyanoborohydride (718 mg, 11.4 mmol) and acetic acid (5 mL) was added to the mixture. The resultant mixture was stirred at RT for 16 h. The mixture was diluted with EtOAc (100 mL), washed with water (50 mL) and brine, dried over Na2S04, filtered and concentrated and purified by silica gel column, eluting with PE:EA (5: 1) to get title compound ethyl 2-chloro- 5-((3,5-dimethoxyphenylamino)methyl)-N-ethylpyridin-4-amine (1.1 g, 85%) as a white solid. MS (ES+) C16H20C1N3O2 requires: 321, found: 322 [M + H]+.
Step 5: Synthesis of 7-chloro-3-(3,5-dimethoxyphenyl)- l-ethyl-3,4-dihydropyrido[4,3- d]pyrimidin-2(lH)-one
Figure imgf000052_0001
To a solution of triphosgene (345 mg, 1.2 mmol) in anhydrous THF (30 mL) at 0 °C was added 2-chloro-5-((3,5-dimethoxyphenylamino)methyl)-N-ethylpyridin-4-amine (1.1 g, 3.4 mmol) and DIPEA (877 mg, 6.8 mmol). After that, the solution was stirred at RT for 16 h. Then the solution was concentrated and purified by silica gel column, eluting with DCM:MeOH from 50: 1 to 25: 1 to give title compound 7-chloro-3-(3,5-dimethoxyphenyl)-l-ethyl-3,4- dihydropyrido[4,3-d]pyrimidin-2(lH)-one (250 mg, 30%) as a white solid. MS (ES+)
C17H18C1N303 requires: 347, found: 348 [M+H]+.
Step 6: Synthesis of 7-chloro-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-ethyl-3,4- dihydropyrido[4,3-d]pyrimidin-2(lH)-one
Figure imgf000052_0002
To a solution of 7-chloro-3-(3,5-dimethoxyphenyl)- l-ethyl-3,4-dihydropyrido[4,3- d]pyrimidin-2(lH)-one (250 mg, 0.72 mmol) in THF (10 mL) at -70 °C was dropwise added a solution of sulfuryl dichloride (243 mg, 1.8 mmol) in THF (2 mL). After addition, the mixture was stirred at -20 °C for another 0.5 h. LCMS showed the reaction was completed. The reaction was quenched with water (0.1 mL), and the solvents were removed under reduced pressure. The precipitate was washed with MeOH, and dried to give 7-chloro-3-(2,6-dichloro-3,5- dimethoxyphenyl)-l-ethyl-3,4-dihydropyrido[4,3-d]pyrimidin-2(lH)-one.
Synthetic Example 4: Synthesis of 6-(2,6-dichloro-3,5-dimethoxyphenyl)-8-methyl-2- (methylsulfonyl)pyridor2,3-dlpyrimidin-7(8H)-one
Step 1: Synthesis of eth l 4-(methylamino)-2-(methylthio)pyrimidine-5-carboxylate
Figure imgf000053_0001
A mixture of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (5.0 g, 21.5 mmol) and 29% methylamine (5.75 g, 53.72 mmol, methanol (MeOH) solution) in tetrahydrofuran (THF) (100 mL) was stirred at room temperature for 2 hours. The reaction mixture was then concentrated, followed by the addition of sodium bicarbonate (NaHC03) (aq., 20 mL), and the resulting solution was extracted with ethyl acetate (EtOAc) (3 x 50 mL). The combined organic layers were washed with water and brine, dried over sodium sulfate, filtered, and concentrated to afford ethyl 4-(methylamino)-2-(methylthio)pyrimidine-5-carboxylate (4.68 g, 96%) as a yellowish solid. MS (ES+) C9Hi3N302S requires: 227, found: 228 [M + H]+.
Step 2: Synthesis of (4-(methylamino)-2-(methylthio)pyrimidin-5-yl)methanol
Figure imgf000053_0002
To a suspension of lithium aluminum hydride (LiAlH4) (1.140 g, 30 mmol) in THF (100 mL) was added ethyl 4-(methylamino)-2-(methylthio)pyrimidine-5-carboxylate (4.536 g, 20 mmol), and the reaction mixture was stirred at room temperature for 2 hours. The solution was carefully quenched with H20 (2 mL), sodium hydroxide (NaOH) (aq., 15%, 2 mL) and additional H20 (7 mL), and then stirred for 1 hour. The mixture was extracted with EtOAc (2 x 100 mL), and the combined organic layers were washed with water and brine, dried over sodium sulfate, and concentrated to give (4-(methylamino)-2-(methylthio)pyrimidin-5-yl)methanol (3.2 g, 85%) as a yellowish solid. MS (ES+) C7HuN3OS requires: 185, found: 186 [M + H]+. Step 3: Synthesis of 4-(methylamino)-2-(methylthio)pyrimidine-5-carbaldehyde
Figure imgf000054_0001
A suspension of (4-(methylamino)-2-(methylthio)pyrimidin-5-yl)methanol (3.1 g, 16.73 mmol) and manganese dioxide (7.27 g, 83.67 mmol) in DCM (40 mL) was stirred at room temperature for 12 hours. The resulting precipitate was filtered off, and the filtrate was concentrated to give 4-(methylamino)-2-(methylthio)pyrimidine-5-carbaldehyde (2.8 g, 91%) as a yellowish solid. MS (ES+) C7H9N3OS requires: 183, found: 184 [M + H]+.
Step 4: Synthesis of methyl 2-(3,5-dimethoxyphenyl)acetate
Figure imgf000054_0002
To a solution of 2-(3,5-dimethoxyphenyl)acetic acid (5) (600 mg, 3.06 mmol) in MeOH (30 mL) was added dropwise thionyl chloride (3 mL) at 0 °C, and the reaction mixture was stirred at room temperature overnight. The reaction was monitored by liquid chromatography- mass spectrometry (LCMS). The mixture was diluted with saturated sodium bicarbonate (aq., 20 mL) and extracted by EtOAc (3 x 20 mL). The combined organic layers were washed with water and brine, dried over sodium sulfate, filtered and concentrated to give methyl 2-(3,5- dimethoxyphenyl) acetate (crude, 700 mg) as a yellow oil. MS (ES+) C11H1404 requires: 210, found: 211 [M + H] +.
Step 5: Synthesis of 6-(3,5-dimethoxyphenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin- 7(8H)-o
Figure imgf000054_0003
A solution of 2-(3,5-dimethoxyphenyl)acetate (6) (440 mg, 2.40 mmol), 4-amino-2- (methylthio)pyrimidine-5-carbaldehyde (4) (605 mg, 2.88 mmol) and potassium carbonate (662 mg, 4.8 mmol) in DMF (30 mL) was stirred at 110 °C for 3 hours. The reaction was monitored by LCMS. The reaction mixture was diluted with H20 (30 mL), and extracted by EtOAc (3 x 40 mL). The combined organic layers were washed with water and brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, petroleum ether/EtOAc = 2: 1) to afford 6-(3,5-dimethoxyphenyl)-8-methyl-2-
(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (7) (683 mg, 83%) as a white solid. MS (ES+) CnHnNsOsS requires: 343, found: 344 [M + H]+.
Step 6: Synthesis of 6-(3,5-dimethoxyphenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3- d]pyrimidin-7(8H)-one
Figure imgf000055_0001
To a solution of 6-(3,5-dimethoxyphenyl)-8-methyl-2- (methylthio)pyrido[2,3- d]pyrimidin-7(8H)-one (1.05 g, 3.1 mmol) in methanol/dichloromethane (MeOH/DCM) (20 mL/20 mL) was added a solution of Oxone® (potassium peroxymonosulfate) (11.3 g, 18.4 mmol) in H20 (20 mL) at room temperature, and the reaction mixture was stirred at 40 °C for 18 hours. The reaction was monitored by LCMS. The reaction mixture was diluted with H20/DCM (150 mL/100 mL), and the aqueous phase was extracted with DCM (100 mL). The combined organic layers were washed with water (200 mL) and brine (200 mL), dried over sodium sulfate, filtered, and concentrated. The crude product was recrystallizated with EtOAc to afford 6-(3,5- dimethoxyphenyl)-8-methyl-2- (methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (8) (910 mg, yield 78%) as yellow solid. MS (ES+) CnHnNsOsS, requires: 375, found: 376 [M + H]+.
Step 7: Synthesis of 6-(2,6-dichloro-3,5-dimethoxyphenyl)-8-methyl-2- (methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one
Figure imgf000056_0001
To a solution of 6-(3,5-dimethoxyphenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3- d]pyrimidin-7(8H)-one (8) (938 mg, 2.5 mmol) in acetonitrile (50 mL) was slowly added a solution of sulfuryl chloride (1.34 g, 10.0 mmol) in acetonitrile (25 mL) over a period of 0.5 hour at a temperature ranging from -10 °C to 0 °C. The reaction was monitored by thin layer chromatography (TLC). The reaction mixture was quenched by adding H20 (10 mL). The resultant reaction solution was concentrated under reduced pressure, and the residue was recrystallizated with EtO Ac/petroleum ether = 1:2 to give 6-(2,6-dichloro-3,5-dimethoxyphenyl) -8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (9) (760 mg, 69% yield) as yellow solid. MS (ES+) CnHisClaNsOsS requires: 443, 445, found: 444, 446 [M + H]+.
Synthesis of Common Intermediates:
Intermediate A: 7-fluoro-2,2-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4- dihydro-2H-benzo[b] [ 1 ,4] oxazine
Step 1: Synthesis of 6-bromo-7-fluoro- -dimethyl-2H-benzo[b][l,4]oxazin-3(4H)-one
Figure imgf000056_0002
2-amino-4-bromo-5-fluorophenol hydrochloride (1.0 g, 4.12 mmol) was taken up in dichloroethane (10 ml) and 2-bromo-2-methylpropanoyl bromide (0.692 ml, 4.54 mmol) was added, followed by addition of DIEA (0.720 ml, 4.12 mmol). Stirred at room temperature for 3 hours. Reaction was cooled to OoC and water added, stirred at OoC for 10 minutes. Layers were separated and solvent removed. Residue was then taken back up in acetonitrile and K2C03(1.65 g, 12 mmol) added. Reaction was then heated to reflux overnight. Complete conversion to desired product. Filtered of solid and removed solvent. The residue was purified via flash chromatography (0-40% Hex/EtOAc; 12g column). Recovered 6-bromo-7-fluoro-2,2-dimethyl- 2H-benzo[b][l,4]oxazin-3(4H)-one (0.270g, 24% yield) as a pink solid. LCMS M+H: 275 Step 2: Synthesis of 6-bromo-7-fluoro-2,2-dimethyl-3,4-dihydro-2H-benzo[b][l,4]oxazine
Figure imgf000057_0001
6-bromo-7-fluoro-2,2-dimethyl-2H-benzo[b][l,4]oxazin-3(4H)-one (0.270 g, 0.985 mmol) was taken up in THF (5 ml) and BH3.DMS (1.970 ml, 3.94 mmol) was added slowly at room temperature. The reaction was heated to reflux for 2 hours. Reaction was removed from oil bath and cooled to room temperature and quenched slowly with Methanol(lml). Mixture was stirred overnight at room temperature. Diluted with DCM and water, separated layers and removed solvent to give title compound, (0.21 lg, 82% yield), that was carried on without purification. LCMS M+H: 261
Step 3: Synthesis of 7-fluoro-2,2-dimethyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4- dihydro-2H-benzo[b] [ 1 ,4] oxazine
Figure imgf000057_0002
6-bromo-7-fluoro-2,2-dimethyl-3,4-dihydro-2H-benzo[b][l,4]oxazine (0.210 g, 0.807 mmol), BIS(PINACOLATO)DIBORON (0.410 g, 1.615 mmol), PdC12(dppf)-CH2C12Adduct (0.066 g, 0.081 mmol), and POTASSIUM ACETATE (0.238 g, 2.422 mmol) were sealed in a 20ml reaction tube. The tube was evacuated and purged with N2- Addition of Dioxane (8.07 ml) followed purging with N2- Reaction was heated to 100 °C overnight. Cooled to room temperature and partitioned between EtO Ac/water, filtered through celite. Layers were separated and aqueous layer extracted with EtOAcx2. Combined organics were washed with brinex2. Dried over Na2S04 and solvent removed. Recovered 7-fluoro-2,2-dimethyl-6-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4-dihydro-2H-benzo[b][l,4]oxazine (0.147 g, 0.479 mmol, 59.3 % yield) crude was carried on as is. LCMS M+H: 308. Intermediate B: 7-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4-dihyd] benzo [b] [ 1 ,4] oxazine
Step 1: Synthesis of 7-methyl-2H-benzo[b][l,4]oxazin-3(4H)-one
Figure imgf000058_0001
To a solution of 2-amino-5-methyl-phenol (3.0 g, 24.4 mmol) in acetonitrile (203 mL), 2- chloroacetyl chloride (2.75 g, 24.4 mmol) was added dropwise over 20 minutes, followed by potassium carbonate (10.1 g, 73.1 mmol). The reaction mixture was heated at reflux for 2 hours, then cooled to room temperature. The mixture was filtered and the solid was washed with 50 mL of acetonitrile. The filtrate was concentrated under vacuum. The residue was dissolved into dichloromethane (200 mL), washed with water (100 mL), dried over sodium sulfate, filtered, and concentrated under vacuum to yield 7-methyl-2H-benzo[b][l,4]oxazin-3(4H)-one (1.6 g, 40% yield) as a beige/brown solid MS: M+l = 164.1.
Step 2: Synthesis of 6-bromo-7-methyl-2H-benzo[b][l,4]oxazin-3(4H)-one
Figure imgf000058_0002
To a stirred suspension of 7-methyl-2H-benzo[b][l,4]oxazin-3(4H)-one (1.065 g, 6.53 mmol) in carbon tetrachloride (22 mL) was added dropwise over 20-25 minutes a solution of bromine (1.046 g, 6.55 mmol) in carbon tetrachloride (14 mL). The suspension was stirred over 5 hours. The reaction progressed slowly to nearly 70% completion. The mixture was then filtered. The solid was washed with carbon tetrachloride followed by DCM. The filtrate was concentrated down and purified by silica gel chromatography to yield were collected, concentrated down, and dried to yield 6-bromo-7-methyl-2H-benzo[b][l,4]oxazin-3(4H)-one (0.42 g, 27% yield). MS: M+l = 242.0.
Step 3: Synthesis of 6-bromo-7-methyl-3,4-dihydro-2H-benzo[b][l,4]oxazine
Figure imgf000058_0003
6-Bromo-7-methyl-2H-benzo[b][l,4]oxazin-3(4H)-one (84.6 mg, 0.349 mmol) was stirred at 0°C in tetrahydrofuran (2 mL), and borane-methyl sulfide complex (0.35 mL of 2N in
THFsolution, 0.701 mmol) was added slowly over 5 minutes. The reaction was warmed to room temperature and then stirred at 70-75°C for nearly 2 hours. The reaction was quenched by cooling the clear solution back to 0°C and slowly adding IN aqueous HC1 (0.2 mL). The solution was then stirred 1 hour at 70°C. After cooling to room temperature, the solution was basified to pH 8-9 with aqueous IN NaOH, diluted with water (~8 mL), and extracted with EtOAc (-20 mL). The organic extract was dried over sodium sulfate, filtered, concentrated down, and dried to yield 6-bromo-7-methyl-3,4-dihydro-2H-benzo[b][l,4]oxazine (89 mg, 90% purity, 100% yield). MS: M+l = 228.0.
Step 4: Synthesis of 7-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4-dihydro-2H- benzo [b] [ 1 ,4] oxazine
Figure imgf000059_0001
A mixture of 6-bromo-7-methyl-3,4-dihydro-2H-benzo[b][l,4]oxazine (89 mg, 90% purity, 0.351 mmol), bis(pinacolato)diboron (134 mg, 0.526 mmol), and potassium acetate (103 mg, 1.05 mmol) in 1,4-dioxane (3.5 mL) was degassed with nitrogen for 5 min at RT and heated at 100°C under a nitrogen atmosphere. After 30 min, dichloro[l,l'-bis(diphenylphosphino)- ferrocene]palladium (II) DCM adduct (28.6 mg, 0.035 mmol) was added, and the reaction mixture was stirred at 100°C. After 18 nours, the reaction was cooled to room temperature and filtered with EtOAc through a celite plug. The filtrate was concentrated down and partially purified by silica gel chromatography to yield 7-methyl-6-(4,4,5,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)-3,4-dihydro-2H-benzo[b][l,4]oxazine (137.8 mg, 70% purity, 100% yield) as a light amber oil. MS: M+l= 276.2.
Intermediate C: 7-fluoro-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4-dihydro-2H- benzo[b][ 1,4] oxazine
Figure imgf000059_0002
7-Fluoro-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4-dihydro-2H-benzo[b][l,4]oxazine was prepared in similar manner as 7-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4- dihydro-2H-benzo[b][l,4]oxazine from commercially available 6-bromo-7-fluoro-3,4-dihydro- 2H-benzo[b][l,4]oxazine. MS: M+l = 280.1.
Intermediate D: 7-fluoro-3-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4-dihydro- 2H-benzo[b] [ 1 ,4] oxazine
Step 1: Synthesis of 6-bromo-7-fluoro-3-meth l-2H-benzo[b][l,4]oxazine
Figure imgf000060_0001
To a stirred 0°C solution of 2-amino-4-bromo-5-fluorophenol (713 mg, 2.94 mmol) in acetone (2 mL) were added chloroacetone (299 mg, 3.23 mmol) and potassium carbonate (447 mg, 3.23 mmol). The mixture was warmed to room temperature and stirred 16 hours. A white solid was removed by filtration. The filtrate was concentrated down and dried to yield 6-bromo-7-fluoro- 3-methyl-2H-benzo[b][l,4]oxazine (660 mg, 92% yield) as a rust-colored solid. MS: M+l =
243.9.
Step 2: Synthesis of 6-bromo-7-fluoro-3-methyl-3,4-dihydro-2H-benzo[b][l,4]oxazine
Figure imgf000060_0002
To a stirred mixture of 6-bromo-7-fluoro-3-methyl-2H-benzo[b][l,4]oxazine (660 mg, 2.70 mmol) in trifluoroacetic acid (10 mL) was added in small portions sodium cyanoborohydride (170 mg, 2.70 mmol)). After addition, the mixture was stirred about 3 hours. The mixture was slowly added to 150 mL of 2N aqueous NaOH and extracted with ethyl acetate (2 x 100 mL). The organic layers were washed with water, combined, dried over sodium sulfate, filtered, concentrated down, and dried to yield 6-bromo-7-fluoro-3-methyl-3,4-dihydro-2H- benzo[b][ 1,4] oxazine (602 mg, 83% yield) as a brown, thick oil. MS: M+l = 247.9.
Step 3: Synthesis of 7-fluoro-3-methyl-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,4- dihydro-2H-benzo[b] [ 1 ,4] oxazine
Figure imgf000060_0003
7-Fluoro-3-methyl-6-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-3,4-dihydro-2H- benzo[b] [l,4]oxazine was prepared in similar manner as 7-methyl-6-(4,4,5,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)-3,4-dihydro-2H-benzo[b] [l,4]oxazine from -bromo-7-fluoro-3-methyl-3,4- dihydro-2H-benzo[b] [l,4]oxazine. MS: M+l = 294.1.
General Protocol for Coupling Reaction
1) Pd mediated
coupling reaction
2) Leaving group
Figure imgf000061_0001
formation
Figure imgf000061_0002
Aromatic bicycle (E) (LG1 can be e.g. , CI, Br, or I; and P can be hydrogen or a protecting group) can be reacted with a boron, tin or zinc aryl or heteroaryl amine (D) (M can be -B(OR)2 where R is hydrogen or alkyl, -Sn(alkyl)3, or -Zn-halo) via a palladium-mediated coupling reaction, e.g. , Suzuki, Stille, or Negishi coupling, to provide intermediate (E) with a new carbon- carbon bond formed. The amino group of intermediate (E) can be reacted with a carboxylic acid, or an ester, e.g. , an activated ester, such as an N-hydroxysuccinimide ester, or an acyl chloride, to provide compounds of Formula I. 1H NMR and LCMS data for Compounds 1 to 56 is summarized below. The compounds below can be synthesized using procedures analogous to the Synthetic Examples noted in the table.
Figure imgf000062_0001
Figure imgf000063_0001
!H NMR (400 MHz, DMSO-i ) δ 12.99 (s, IH), 9.63
(s, IH), 8.67 (dt, J = 8.4, 2.3 Hz, 2H), 8.01 (s, IH),
7.61 (s, IH), 7.59 - 7.53 (m, IH), 7.37 - 7.23 (m, IH), 511
1
7.03 (s, IH), 6.64 (dd, J = 16.9, 10.3 Hz, IH), 6.36
(dd, J = 17.0, 1.5 Hz, IH), 5.89 (dd, J = 10.2, 1.5 Hz,
IH), 3.98 (d, J = 1.8 Hz, 6H), 1.06 (s, 3H).
:H NMR (400 MHz, DMSO-i ) δ 9.69 (s, IH), 8.31
(dd, J = 8.7, 2.1 Hz, IH), 8.08 (d, J = 8.7 Hz, IH),
8.03 (d, J = 1.9 Hz, IH), 7.82 (dd, J = 8.6, 2.0 Hz,
523
1 IH), 7.11 (d, J = 8.6 Hz, IH), 7.05 (s, IH), 6.88 (dd, J
= 16.7, 10.4 Hz, IH), 6.35 (dd, J = 16.8, 2.1 Hz, IH),
5.89 (dd, J = 10.4, 2.1 Hz, IH), 4.39 (t, J = 4.5 Hz,
2H), 4.03 (d, J = 4.7 Hz, 2H), 3.99 (s, 6H).
2
1 524
:H NMR (400 MHz, DMSO-i ) δ 11.25 (s, IH), 9.07
(s, IH), 8.22 (d, = 8.4 Hz, IH), 8.09 (s, IH), 7.91 (d,
= 7.5 Hz, IH), 7.82 (s, IH), 7.49 (t, = 7.7 Hz, IH),
2 7.32 (t, = 7.6 Hz, IH), 7.02 (s, IH), 6.43 (dd, = 524 17.0, 10.2 Hz, IH), 6.22 (dd, = 17.0, 1.7 Hz, IH),
5.74 (dd, = 10.1, 1.8 Hz, IH), 4.37 (q, = 7.9, 7.5
Hz, 2H), 3.97 (s, 6H), 1.23 (t, = 7.0 Hz, 3H).
:H NMR (400 MHz, DMSO-d6) δ 10.34 (s, IH), 9.02
(s, IH), 8.46 (t, J = 2.0 Hz, IH), 8.05 (s, IH), 8.02 - 7.87 (m, 3H), 7.50 (t, J = 7.9 Hz, IH), 7.01 (s, IH),
2 524 6.48 (dd, J = 17.0, 10.1 Hz, IH), 6.29 (dd, J = 16.9,
2.0 Hz, IH), 5.78 (dd, J = 10.1, 2.1 Hz, IH), 4.43 (q, J
= 7.0 Hz, 2H), 3.97 (s, 6H), 1.27 (t, J = 7.1 Hz, 3H).
!H NMR (400 MHz, DMSO-d6) δ 10.38 (s, IH), 9.06
(s, IH), 8.31 (dd, J = 7.1, 2.8 Hz, IH), 8.18 (s, IH),
7.97 - 7.82 (m, 2H), 7.37 (dd, J = 11.0, 8.9 Hz, IH),
1 7.06 (d, J = 7.8 Hz, IH), 6.44 (dd, J = 17.0, 10.0 Hz, 526 IH), 6.28 (dd, J = 17.0, 2.1 Hz, IH), 5.78 (dd, J =
10.0, 2.1 Hz, IH), 4.33 (q, J = 7.0 Hz, 2H), 3.94 (s,
3H), 3.93 (s, 3H), 1.27 (t, J = 7.0 Hz, 3H).
!H NMR (400 MHz, DMSO-d6) δ 10.29 (s, IH), 8.38
(s, IH), 8.32 (s, IH), 7.87 (d, J = 8.1 Hz, IH), 7.80 (d,
J = 7.8 Hz, IH), 7.50 - 7.39 (m, 2H), 6.99 (s, IH),
3 6.46 (dd, J = 16.9, 10.1 Hz, IH), 6.28 (dd, J = 17.0, 527 2.0 Hz, IH), 5.77 (dd, J = 10.1, 2.1 Hz, IH), 4.71 (s,
2H), 4.01 (q, J = 7.2 Hz, 2H), 3.96 (s, 6H), 1.21 (t, J =
6.9 Hz, 3H).
Figure imgf000065_0001
!H NMR (400 MHz, DMSO-d6) δ 10.40 (s, IH), 9.27
(s, IH), 8.56 (dd, J = 7.0, 2.8 Hz, IH), 8.08 (s, IH),
7.93 (dt, J = 9.0, 3.4 Hz, IH), 7.37 (dd, J = 10.9, 8.9
4 Hz, IH), 7.03 (s, IH), 6.44 (dd, J = 17.0, 10.1 Hz, 543 IH), 6.29 (dd, J = 17.0, 2.0 Hz, IH), 5.79 (dd, J =
10.1, 2.1 Hz, IH), 4.48 (q, J = 6.9 Hz, 2H), 3.97 (s,
6H), 1.29 (t, J = 7.0 Hz, 3H).
!H NMR (400 MHz, DMSO-d6) δ 10.32 (s, IH), 8.42
(s, IH), 8.19 (dd, J = 7.2, 2.7 Hz, IH), 7.90 - 7.79 (m,
IH), 7.39 (s, IH), 7.32 (dd, J = 10.9, 8.9 Hz, IH), 7.00
3 (s, IH), 6.42 (dd, J = 17.0, 10.1 Hz, IH), 6.27 (dd, J = 545 17.0, 2.1 Hz, IH), 5.77 (dd, J = 10.0, 2.1 Hz, IH),
4.73 (s, 2H), 3.96 (s, 6H), 3.95 - 3.87 (m, 2H), 1.19 (t,
J = 7.0 Hz, 3H).
1 548
IH NMR (400 MHz, DMS0-d6) δ 9.69 (s, IH), 8.31
(dd, J = 8.6, 2.1 Hz, IH), 8.08 (d, J = 8.7 Hz, IH),
8.03 (d, J = 1.9 Hz, IH), 7.82 (dd, J = 8.7, 2.0 Hz,
1 IH), 7.06 (d, J = 7.9 Hz, 2H), 6.95 (dd, J = 16.7, 10.4 551 Hz, IH), 6.39 (dd, J = 16.6, 2.1 Hz, IH), 5.91 (dd, J =
10.3, 2.1 Hz, IH), 3.99 (s, 6H), 3.86 (s, 2H), 1.32 (s,
6H).
!H NMR (400 MHz, DMS0-d6) δ 9.04 (s, IH), 9.01
(s, IH), 8.04 (s, IH), 7.95 - 7.85 (m, 2H), 7.41 (d, J =
7.8 Hz, IH), 7.01 (s, IH), 6.79 (dd, J = 16.5, 9.9 Hz,
2 IH), 6.34 (dd, J = 16.7, 1.9 Hz, IH), 5.85 (dd, J = 550 10.4, 2.2 Hz, IH), 4.42 (q, J = 7.0 Hz, 2H), 4.29 (t, J =
8.4 Hz, 2H), 3.97 (s, 6H), 3.24 (t, J = 8.3 Hz, 2H),
1.27 (t, J = 7.0 Hz, 3H).
!H NMR (400 MHz, DMSO- ) δ 9.47 (s, IH), 8.29
(dd, = 8.6, 2.1 Hz, IH), 7.87 (s, IH), 7.48 (s, IH),
7.09 (d, = 8.6 Hz, IH), 7.00 (s, IH), 6.88 (dd, =
553
1 16.9, 10.5 Hz, IH), 6.35 (dd, = 16.7, 2.1 Hz, IH),
5.88 (dd, = 10.4, 2.1 Hz, IH), 4.38 (t, = 4.6 Hz,
2H), 4.02 (t, = 4.7 Hz, 2H), 3.97 (s, 6H), 3.92 (s,
3H).
!H NMR (400 MHz, DMS0-d6) δ 8.95 (s, IH), 8.41 - 8.28 (m, IH), 8.11 - 7.93 (m, 2H), 7.85 (s, IH), 7.07
(d, J = 8.6 Hz, IH), 7.01 (s, IH), 6.85 (dd, J = 16.8,
2 552 10.4 Hz, IH), 6.32 (dd, J = 16.8, 2.1 Hz, IH), 5.85
(dd, J = 10.4, 2.1 Hz, IH), 4.37 (t, J = 4.6 Hz, 4H),
4.00 (t, J = 4.6 Hz, 4H), 3.96 (s, 6H), 3.74 (s, 3H).
:H NMR (300 MHz, DMS0-d6) δ 9.73 (s, IH), 8.12
(d, J = 8.7 Hz, IH), 8.08 (d, J = 1.8 Hz, IH), 7.87 (dd,
J = 8.7, 1.9 Hz, IH), 7.07 (s, IH), 7.03 (d, J = 11.7 Hz,
1 IH), 6.93 - 6.77 (m, 2H), 6.32 (dd, J = 16.7, 1.9 Hz, 554 IH), 5.87 (dd, J = 10.4, 1.8 Hz, IH), 4.45 - 4.20 (m,
IH), 4.00 (s, 6H), 3.48 - 3.36 (m, 2H), 1.21 (d, J = 7.0
Hz, 3H). IH NMR (400 MHz, DMSO-d6) δ 10.19 (s, IH), 9.01
(s, IH), 8.18 (d, J = 2.7 Hz, IH), 8.05 (s, IH), 8.03 (s,
IH) 7.88 (dd, J = 8.9, 2.8 Hz, IH), 7.19 (d, J = 9.0 Hz,
2 IH), 7.01 (s, IH), 6.43 (dd, J = 16.9, 10.1 Hz, IH), 554 6.24 (dd, J = 17.0, 2.1 Hz, IH), 5.73 (dd, J = 10.2 Hz,
IH), 4.31 (q, J = 7.1 Hz, 2H), 3.97 (s, 6H), 3.89 (s,
3H), 1.29 (t, J = 7.1 Hz, 3H).
!H NMR (400 MHz, DMSO-d6) δ 10.38 (s, IH), 9.04
(s, IH), 8.37 - 8.25 (m, IH), 8.04 (d, J = 10.4 Hz,
IH), 7.96 - 7.85 (m, IH), 7.37 (dd, J = 11.1, 8.9 Hz,
2 IH), 7.01 (s, IH), 6.44 (dd, J = 17.0, 10.1 Hz, IH), 556 6.28 (dd, J = 17.0, 2.0 Hz, IH), 5.78 (dd, J = 10.1, 2.1
Hz, IH), 5.30-5.17 (m, IH), 3.97 (s, 6H), 1.56 (d, J =
6.9 Hz, 6H).
!H NMR (400 MHz, DMSO-d6) δ 10.56 (s, IH), 9.08
(s, IH), 8.12 - 8.03 (m, 2H), 7.98-7.95 (m, 2H), 7.02
(s, IH), 6.42 (dd, J = 17.0, 9.9 Hz, IH), 6.30 (dd, J =
2 560 17.0, 2.1 Hz, IH), 5.81 (dd, J = 9.9, 2.1 Hz, IH), 4.34
(q, J = 6.8 Hz, 2H), 3.97 (s, 6H), 1.26 (t, J = 7.0 Hz,
3H).
:H NMR (400 MHz, DMSO-i ) δ 11.03 (s, IH), 9.04
(s, IH), 8.16 (d, = 8.2 Hz, IH), 8.05 (s, IH), 7.96 - 7.77 (m, 2H), 7.48 (t, = 7.9 Hz, IH), 7.32 (t, = 7.6
Hz, IH), 7.01 (s, IH), 6.42 (dd, = 17.0, 10.2 Hz,
2 565 IH), 6.22 (d, = 16.8 Hz, IH), 5.74 (d, = 9.5 Hz,
IH), 5.44 (s, IH), 3.96 (s, 6H), 2.09 (s, 2H), 1.95 (d,
= 21.5 Hz, 3H), 1.62 (s, 2H), 1.19 (d, = 26.6 Hz,
IH).
lU NMR (400 MHz, DMSO- ) δ 10.34 (s, IH), 8.99
(s, IH), 8.45 (s, IH), 8.11 (s, IH), 8.01 (s, IH), 7.98 - 7.87 (m, 2H), 7.50 (t, = 7.9 Hz, IH), 7.01 (s, IH),
2 6.48 (dd, = 16.9, 9.8 Hz, IH), 6.37 - 6.18 (m, IH), 565 5.89 - 5.69 (m, IH), 5.54 (d, = 10.0 Hz, IH), 3.96
(s, 6H), 2.13 (s, 2H), 1.99 (s, 3H), 1.67 (s, 2H), 1.23
(s, IH).
lU NMR (400 MHz, DMSO-d6) δ 9.70 (s, IH), 8.16 - 7.99 (m, 2H), 7.83 (dd, J = 8.6, 2.0 Hz, IH), 7.06 (s,
1 IH), 6.88 (s, IH), 6.39 - 6.27 (m, IH), 5.85 (dd, J = 565 10.4, 2.1 Hz, IH), 3.99 (s, 8H), 3.82 (s, 2H), 2.62 (s,
3H), 1.31 (s, 6H).
!H NMR (400 MHz, DMSO-i ) δ 8.98 (s, IH), 8.03
(d, = 8.5 Hz, 3H), 7.92 (s, IH), 7.39 (d, = 7.9 Hz,
IH), 7.01 (s, IH), 6.63 (dd, = 16.7, 10.3 Hz, IH),
2 6.31 - 6.21 (m, IH), 5.77 - 5.67 (m, IH), 4.42 (q, = 564 7.0 Hz, 2H), 3.96 (d, = 0.8 Hz, 6H), 3.80 (t, = 6.5
Hz, 2H), 2.79 (t, = 6.5 Hz, 2H), 1.97 - 1.89 (m, 2H),
1.24 (t, = 7.0 Hz, 3H). !H NMR (400 MHz, DMSO- ) δ 11.71 (s, IH), 9.28
(d, / = 1.0 Hz, IH), 8.44 (d, / = 8.3 Hz, IH), 8.29 (dd,
/ = 8.0, 1.6 Hz, IH), 8.08 (d, / = 1.1 Hz, IH), 7.63 - 7.46 (m, IH), 7.39 - 7.22 (m, IH), 7.02 (s, IH), 6.50
4 566 (dd, / = 17.0, 10.2 Hz, IH), 6.27 (dd, / = 17.0, 1.6 Hz,
IH), 6.05 (q, / = 8.7 Hz, IH), 5.79 (dd, / = 10.2, 1.6
Hz, IH), 3.97 (s, 6H), 2.19 (d, / = 12.5 Hz, 2H), 1.93
(d, / = 19.0 Hz, 2H), 1.62 (s, 2H).
!H NMR (400 MHz, DMSO-d6) δ 8.95 (s, IH), 8.30
(b.s., IH), 8.04 - 7.98 (m, 2H), 7.87 (s, IH), 7.07 (d, J
= 8.6 Hz, IH), 7.01 (s, IH), 6.85 (dd, J = 16.7, 10.4
2 566 Hz, IH), 6.32 (dd, J = 16.7, 2.1 Hz, IH), 5.84 (dd, J =
10.4, 2.1 Hz, IH), 4.47 - 4.33 (m, 4H), 4.00 (t, J = 4.7
Hz, 2H), 3.96 (s, 6H), 1.24 (t, J = 7.0 Hz, 3H).
lU NMR (400 MHz, DMSO-d6) δ 9.72 (s, IH), 8.15 - 8.01 (m, 2H), 7.85 (dd, J = 8.7, 2.0 Hz, IH), 7.06 (s,
1 IH), 6.94 (d, J = 11.8 Hz, IH), 6.35 (dd, J = 16.7, 2.1 569 Hz, IH), 5.89 (dd, J = 10.4, 2.1 Hz, IH), 3.99 (s, 8H),
3.87 (s, 2H), 1.33 (s, 6H).
IH NMR (400 MHz, DMSO-d6) δ 10.32 (s, IH), 8.99
(s, IH), 8.04 (s, IH), 7.97 (d, J = 1.7 Hz, IH), 7.90 (s,
IH), 7.65 (s, IH), 7.49 (t, J = 1.9 Hz, IH), 7.02 (s,
2 IH), 6.46 (dd, J = 16.9, 10.1 Hz, IH), 6.29 (dd, J = 569 17.0, 2.1 Hz, IH), 5.78 (dd, J = 10.0, 2.1 Hz, IH),
4.70 (p, J = 6.0 Hz, IH), 3.97 (s, 6H), 3.78 (s, 3H),
1.33 (d, J = 6.0 Hz, 6H).
IH NMR (400 MHz, DMSO-d6) δ 8.31 (s, IH), 8.14
(b.s., IH), 7.85 (d, J = 8.2 Hz, IH), 7.38 (s, IH), 7.02
(d, J = 8.6 Hz, IH), 6.99 (s, IH), 6.83 (dd, J = 16.8,
3 569 10.3 Hz, IH), 6.30 (dd, J = 16.7, 2.1 Hz, IH), 5.83
(dd, IH), 4.67 (s, 2H), 4.35 (t, J = 4.5 Hz, 2H), 4.05 - 3.97 (m, 4H), 3.95 (s, 6H), 1.18 (t, J = 6.9 Hz, 3H).
:H NMR (400 MHz, DMSO-i ) δ 10.24 (s, IH), 9.01
(d, / = 2.8 Hz, IH), 8.46 (s, IH), 8.05 (s, IH), 7.97 - 7.77 (m, 3H), 7.48 (t, / = 8.3 Hz, IH), 7.01 (s, IH),
2 6.87 - 6.70 (m, IH), 6.30 (d, / = 15.5 Hz, IH), 4.42 581 (q, = 8.6, 7.0 Hz, 2H), 3.97 (s, 6H), 3.06 (d, = 5.7
Hz, IH), 2.25 (s, IH), 2.18 (s, 6H), 1.27 (t, / = 6.7 Hz,
3H).
2 595
:H NMR (300 MHz, DMSO-d6) δ 9.02 (s, IH), 8.05
(s, IH), 7.83 (s, IH), 7.06 (d, J = 12.2 Hz, IH), 7.02
(s, IH), 6.93 - 6.72 (m, 2H), 6.31 (dd, J = 16.7, 1.7
2 598 Hz, IH), 5.90 - 5.81 (m, IH), 4.44 - 4.17 (m, 3H),
3.98 (s, 6H), 3.48 - 3.36 (m, 2H), 1.26 (t, J = 7.1 Hz,
3H), 1.19 (d, J = 6.8 Hz, 3H). 54 2 607
lU NMR (400 MHz, DMSO- ) δ 9.15 (s, 1H), 8.21
(dd, = 8.6, 2.1 Hz, 1H), 7.98 (s, 1H), 7.11 (d, / = 8.7
Hz, 1H), 7.01 (s, 1H), 6.86 (dd, = 16.7, 10.4 Hz,
55 4 1H), 6.33 (dd, = 16.7, 2.1 Hz, 1H), 6.14 - 5.97 (m, 608
1H), 5.84 (dd, = 10.4, 2.1 Hz, 1H), 4.47 - 4.33 (m,
2H), 4.01 (t, = 4.6 Hz, 2H), 3.96 (s, 6H), 2.28 (d, =
26.3 Hz, 2H), 1.93 (d, = 35.6 Hz, 5H), 1.64 (s, 2H).
!H NMR (400 MHz, DMSO-d6) δ 9.04 (s, 1H), 8.48 - 8.27 (m, 1H), 8.17 (s, 1H), 8.04 (dd, J = 8.6, 2.2 Hz,
1H), 7.13 - 7.03 (m, 1H), 6.85 (dd, J = 16.7, 10.4 Hz,
56 2 1H), 6.32 (dd, J = 16.8, 2.1 Hz, 1H), 5.84 (dd, J = 607
10.4, 2.1 Hz, 1H), 5.58 - 5.41 (m, 1H), 4.38 (t, J = 4.6
Hz, 4H), 4.01 (t, J = 4.5 Hz, 4H), 3.98 (s, 6H), 2.20 - 1.87 (m, 6H), 1.78 - 1.53 (m, 2H).
Biochemical Activity Assessment
In order to assess the activity of chemical compounds against the relevant kinase of interest, the Caliper LifeSciences electrophoretic mobility shift technology platform is utilized. Fluorescently labeled substrate peptide is incubated in the presence of dosed levels of compounds, a set concentration of kinase and of ATP, so that a reflective proportion of the peptide is phosphorylated. At the end of the reaction, the mix of phosphorylated (product) and non-phosphorylated (substrate) peptides are passed through the microfluidic system of the Caliper LabChip® EZ Reader II, under an applied potential difference. The presence of the phosphate group on the product peptide provides a difference in mass and charge between the product peptide and the substrate peptide, resulting in a separation of the substrate and product pools in the sample. As the pools pass the LEDS within the instrument, these pools are detected and resolved as separate peaks. The ratio between these peaks therefore reflects the activity of the chemical matter at that concentration in that well, under those conditions.
FGFR-4 wild type assay at Km: In each well of a 384-well plate, 0.5 ng/ul of wild type
FGFR-4 (Carna Biosciences, Inc.) was incubated in a total of 12.5 ul of buffer (100 mM HEPES pH 7.5, 0.015% Brij 35, 10 mM MgCl2, ImM DTT) with 1 uM CSKtide (5-FAM- KKKKEEIYFFFG-NH2) and 400 uM ATP at 25 C for 90 minutes in the presence or absence of a dosed concentration series of compound (1% DMSO final concentration). The reaction was stopped by the addition of 70 ul of Stop buffer (100 mM HEPES pH 7.5, 0.015% Brij 35, 35 mM EDTA and 0.2% of Coating Reagent 3 (Caliper Lifesciences)). The plate was then read on a Caliper LabChip® EZ Reader II (protocol settings: -1.9 psi, upstream voltage -700, downstream voltage -3000, post sample sip 35s).
Detection of pMAPK (Thr202/Tyr204) Using Alpha Elisa
MDA-MB453 cells were plated in 96-well cell culture plates at a density of lxlO5 cells.
Cells were allowed to attach, and growth media was replaced with serum free media.
Compounds were added at the indicated concentrations. Following 1 hr incubation in the presence of compound, cells were collected. Cell lysates were prepared and processed according to manufacturer instruction (AlphaScreen® SureFire™ Phospho-ERK 1/2 Kit (Perkin Elmer).
The table below summarizes biochemical data for Compounds 1-56. In the table below, for FGFR4 and pERK alphaLISA: "A" means that the IC50 is less than 10 nM; "B" means the IC50 is greater than or equal to 10 and less than ΙΟΟηΜ; "C" means that the IC50 is greater than or equal to 100 and less than 1000 nM; "D" means that the IC50 is greater than 1000 nM.
Figure imgf000070_0001
16 B
17 A A
18 A B
19 A B
20 A A
21 A A
22 B B
23 A B
24 B B
25 A B
26 B
27 B B
28 A B
29 B
30 A A
31 B
32 A B
33 C
34 B B
35 A B
36 B
37 A
38 B
39 A A
40 A A
41 A
42 A
43 A
44 B 45 A
46 B
47 A B
48 B
49 B
50 A B
51 B
52 A B
53 A B
54 B
55 B
56 A
In the table above: "A" means that the IC50 is less than 10 nM; "B" means the IC50 is greater than or equal to 10 and less than ΙΟΟηΜ; "C" means that the IC50 is greater than or equal to 100 and less than 1000 nM; "D" means that the IC50 is greater than 1000 nM.
Incorporation by Reference
All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference.
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

Claims:
1. A com ound of Formula I, or a pharmaceutically acceptable salt thereof:
Figure imgf000073_0001
Formula I
wherei
Figure imgf000073_0002
denotes a single or double bond;
Warhead is a moiety capable of forming a covalent bond with a nucleophile;
Ring A is a 5-8 membered aryl, 5-12 membered heteroaryl, 3-7 member monocyclic or bicyclic heterocyclyl; or 3-12 membered monocyclic or bicyclic cycloalkyl group;
W is C or N, X and Z are each independently CH or N;
Y is CH or N-R4 where R4 is H, C1-6 alkyl, or 3-12 membered cycloalkyl;
each of R 1 -R 3 is, independently, halo, cyano, optionally substituted Ci_6 alkyl, optionally substituted Ci_6 alkoxy, hydroxy, oxo, amino, amido, alkyl urea, optionally substituted 3-7 member heterocyclyl;
R 7 is hydrogen or C1-6 alkyl; or R 7 together with Ring A forms a 8-12 membered bicyclic heterocyclyl optionally substituted with 1-5 occurrences of R1;
m is 0-5;
n is 0-5; and
p is 0-2.
2. The compound of claim 1, wherein ' denotes a single bond.
3. The compound of claim 1, wherein ' denotes a double bond.
4. The compound of claim 1, wherein one of is a single bond and the other
Figure imgf000074_0001
is a double bond.
5. The compound of claim 1, wherein Ring A is phenyl substituted with 0-3 R1.
6. The compound of claim 1, wherein R is hydrogen or C1-6 alkyl.
7
7. The compound of claim 1, wherein R together with Ring A form an optionally substituted 8-12 membered bicyclic heterocyclyl.
8. The compound of claim 1, wherein m is at least 1 and R1 is selected from the group consisting of halo, optionally substituted C1-6 alkyl, and optionally substituted C1-6 alkoxy.
9. The compound of claim 1, wherein n is 4, with two R 2 being halo and two R 2 being alkoxy.
10. The compound of claim 1, wherein the warhead moiety is selected from the group consisting of
Figure imgf000074_0002
wherein each of Ra, Rb, and Rc is, independently, hydrogen, optionally substituted Q.
optionally substituted 3-12 membered cycloalkyl, or cyano.
11. The compound of claim 1, wherein the warhead moiety is
Figure imgf000074_0003
12. The compound of claim 1, wherein the compound is a compound of Formula 1(a), or a pharmaceutically acceptable salt thereof:
Figure imgf000075_0001
Formula 1(a)
wherein X is CH or N.
13. The compound of claim 1, wherein the compound is a compound of Formula 1(b), or a pharmaceutically acceptable salt thereof:
Figure imgf000075_0002
Formula 1(b) wherein X is CH or N.
14. The compound of claim 1, wherein the compound is a compound of Formula 1(c), or a pharmaceutically acceptable salt thereof:
Figure imgf000075_0003
Formula 1(c)
wherein X is CH or N.
15. The compound of claim 1, wherein the compound is a compound of Formula 1(d), or a pharmaceutically acceptable salt thereof:
Figure imgf000076_0001
Formula 1(d).
16. A pharmaceutical composition comprising a compound of claim 1, and a pharmaceutically acceptable carrier, or a pharmaceutically acceptable salt thereof.
17. A method for treating a condition mediated by FGFR-4, comprising administering to a subject a therapeutically effective amount of a compound of claim 1.
18. A method for treating a condition characterized by overexpression of FGFR-4, comprising administering to a subject a therapeutically effective amount of a compound of claim 1.
19. A method for treating a condition characterized by amplified FGF-19, comprising administering to a subject a therapeutically effective amount of a compound of claim 1.
20. A method for treating a condition characterized by overexpression of FGF-19, comprising administering to a subject a therapeutically effective amount of a compound of claim 1.
21. A method of treating cancer, the method comprising administering to a subject a therapeutically effective amount of compound of claim 1, wherein the cancer is selected from the group consisting of liver cancer, breast cancer, lung cancer, ovarian cancer, or a sarcoma.
22. A method of treating hepatocellular carcinoma, the method comprising administering to a subject a therapeutically effective amount of a compound of claim 1.
23. A method of treating hyperlipidemia, the method comprising administering to a subject a therapeutically effective amount of a compound of claim 1.
24. A compound of claim 1 for use in treating a condition mediated by FGFR-4.
25. A compound of claim 1 for use in treating a condition characterized by overexpression of FGFR-4.
26. A compound of claim 1 for use in treating a condition characterized by amplified FGF-19.
27. A compound of claim 1 for use in treating a condition characterized by overexpression of FGF-19.
28. A compound of claim 1 for use in treating cancer, wherein the cancer is selected from the group consisting of liver cancer, breast cancer, lung cancer, ovarian cancer, or a sarcoma.
29. A compound of claim 1 for use in treating hepatocellular carcinoma.
30. A compound of claim 1 for use in treating hyperlipidemia.
PCT/US2015/011424 2014-01-15 2015-01-14 Heterobicyclic compounds and their use as fgfr4 receptor inhibitors WO2015108992A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461927793P 2014-01-15 2014-01-15
US61/927,793 2014-01-15

Publications (1)

Publication Number Publication Date
WO2015108992A1 true WO2015108992A1 (en) 2015-07-23

Family

ID=52463149

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/011424 WO2015108992A1 (en) 2014-01-15 2015-01-14 Heterobicyclic compounds and their use as fgfr4 receptor inhibitors

Country Status (2)

Country Link
US (2) US9695165B2 (en)
WO (1) WO2015108992A1 (en)

Cited By (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
WO2016134314A1 (en) * 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
WO2016134294A1 (en) * 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
US9434697B2 (en) 2013-10-18 2016-09-06 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
WO2017011776A1 (en) 2015-07-16 2017-01-19 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
WO2017176744A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Methods of treating pediatric cancers
WO2017176751A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
WO2018040885A1 (en) * 2016-09-01 2018-03-08 南京药捷安康生物科技有限公司 Inhibitors of fibroblast growth factor receptor and use thereof
WO2018049233A1 (en) 2016-09-08 2018-03-15 Nicolas Stransky Inhibitors of the fibroblast growth factor receptor in combination with cyclin-dependent kinase inhibitors
WO2018071447A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018071454A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018136661A1 (en) 2017-01-18 2018-07-26 Andrews Steven W SUBSTITUTED PYRAZOLO[1,5-a]PYRAZINE COMPOUNDS AS RET KINASE INHIBITORS
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
WO2018140514A1 (en) * 2017-01-26 2018-08-02 Araxes Pharma Llc 1-(6-(3-hydroxynaphthalen-1-yl)quinazolin-2-yl)azetidin-1-yl)prop-2-en-1-one derivatives and similar compounds as kras g12c inhibitors for the treatment of cancer
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
WO2018170381A1 (en) 2017-03-16 2018-09-20 Andrews Steven W Macrocyclic compounds as ros1 kinase inhibitors
US10111874B2 (en) 2014-09-18 2018-10-30 Araxes Pharma Llc Combination therapies for treatment of cancer
WO2018205916A1 (en) * 2017-05-09 2018-11-15 南京圣和药业股份有限公司 Fgfr4 inhibitor and preparation and use thereof
US10144724B2 (en) 2015-07-22 2018-12-04 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10221154B2 (en) 2013-10-25 2019-03-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10246424B2 (en) 2015-04-10 2019-04-02 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10251889B2 (en) 2009-07-09 2019-04-09 Array BioPharm Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
WO2019075108A1 (en) 2017-10-10 2019-04-18 Metcalf Andrew T Crystalline forms
WO2019075114A1 (en) 2017-10-10 2019-04-18 Mark Reynolds Formulations comprising 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazab icyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
US10273207B2 (en) 2013-03-15 2019-04-30 Araxes Pharma Llc Covalent inhibitors of kras G12C
WO2019080878A1 (en) * 2017-10-24 2019-05-02 Advent Pharma Co. Ltd. Heterocyclics as inhibitors of fibroblast growth factor receptor
US10280172B2 (en) 2016-09-29 2019-05-07 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
CN109745325A (en) * 2017-11-08 2019-05-14 上海翰森生物医药科技有限公司 FGFR4 inhibitor, preparation method and use
WO2019143994A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
WO2019143977A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
CN110092798A (en) * 2018-01-29 2019-08-06 上海新契博生物科技有限公司 It is a kind of as the heterocyclic compound and its synthetic method of FGFR inhibitor and application
US10370386B2 (en) 2013-10-10 2019-08-06 Araxes Pharma Llc Substituted quinolines as inhibitors of KRAS G12C
US10377743B2 (en) 2016-10-07 2019-08-13 Araxes Pharma Llc Inhibitors of RAS and methods of use thereof
US10414757B2 (en) 2015-11-16 2019-09-17 Araxes Pharma Llc 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
US10428064B2 (en) 2015-04-15 2019-10-01 Araxes Pharma Llc Fused-tricyclic inhibitors of KRAS and methods of use thereof
WO2019233438A1 (en) * 2018-06-07 2019-12-12 北京大学深圳研究生院 Kinase selective inhibitor
WO2020028258A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide
US10562888B2 (en) 2015-04-14 2020-02-18 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
WO2020055672A1 (en) 2018-09-10 2020-03-19 Array Biopharma Inc. Fused heterocyclic compounds as ret kinase inhibitors
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
EP3524603A4 (en) * 2016-12-19 2020-04-22 Abbisko Therapeutics Co., Ltd. Fgfr4 inhibitor, preparation method therefor and pharmaceutical use thereof
CN111138459A (en) * 2018-11-06 2020-05-12 南京圣和药业股份有限公司 Optical isomer of FGFR4 inhibitor and application thereof
US10646488B2 (en) 2016-07-13 2020-05-12 Araxes Pharma Llc Conjugates of cereblon binding compounds and G12C mutant KRAS, HRAS or NRAS protein modulating compounds and methods of use thereof
US10647703B2 (en) 2015-09-28 2020-05-12 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
WO2020119606A1 (en) * 2018-12-10 2020-06-18 Guangdong Newopp Biopharmaceuticals Co., Ltd. Heterocyclic compounds as inhibitors of fibroblast growth factor receptor
US10689356B2 (en) 2015-09-28 2020-06-23 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
WO2020131627A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
US10724102B2 (en) 2015-10-26 2020-07-28 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10730867B2 (en) 2015-09-28 2020-08-04 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10736897B2 (en) 2017-05-25 2020-08-11 Araxes Pharma Llc Compounds and methods of use thereof for treatment of cancer
US10745385B2 (en) 2017-05-25 2020-08-18 Araxes Pharma Llc Covalent inhibitors of KRAS
WO2020177534A1 (en) * 2019-03-05 2020-09-10 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10858343B2 (en) 2015-09-28 2020-12-08 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10875842B2 (en) 2015-09-28 2020-12-29 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10882847B2 (en) 2015-09-28 2021-01-05 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
KR20210019510A (en) * 2018-09-14 2021-02-22 아비스코 테라퓨틱스 컴퍼니 리미티드 FGFR inhibitor, its manufacturing method and use
US10975071B2 (en) 2015-09-28 2021-04-13 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
WO2021134004A1 (en) 2019-12-27 2021-07-01 Schrodinger, Inc. Cyclic compounds and methods of using same
US11059819B2 (en) 2017-01-26 2021-07-13 Janssen Biotech, Inc. Fused hetero-hetero bicyclic compounds and methods of use thereof
US11091486B2 (en) 2016-10-26 2021-08-17 Array Biopharma, Inc Process for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11214571B2 (en) 2016-05-18 2022-01-04 Array Biopharma Inc. Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
US11274093B2 (en) 2017-01-26 2022-03-15 Araxes Pharma Llc Fused bicyclic benzoheteroaromatic compounds and methods of use thereof
WO2022055963A1 (en) 2020-09-10 2022-03-17 Schrödinger, Inc. Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer
US11279689B2 (en) 2017-01-26 2022-03-22 Araxes Pharma Llc 1-(3-(6-(3-hydroxynaphthalen-1-yl)benzofuran-2-yl)azetidin-1 yl)prop-2-en-1-one derivatives and similar compounds as KRAS G12C modulators for treating cancer
JP2022523448A (en) * 2019-03-08 2022-04-22 ショウヤオ ホールディングス(ベイジン) カンパニー, リミテッド FGFR4 kinase inhibitor, its production method and use
WO2022087433A1 (en) * 2020-10-23 2022-04-28 Dana-Farber Cancer Institute, Inc. Covalent inhibitors of creatine kinase (ck) and uses thereof for treating and preventing cancer
US11357769B2 (en) 2016-05-10 2022-06-14 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
US11358959B2 (en) 2017-01-26 2022-06-14 Araxes Pharma Llc Benzothiophene and benzothiazole compounds and methods of use thereof
WO2022164789A1 (en) 2021-01-26 2022-08-04 Schrödinger, Inc. Tricyclic compounds useful in the treatment of cancer, autoimmune and inflammatory disoders
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
WO2022197898A1 (en) 2021-03-18 2022-09-22 Schrödinger, Inc. Cyclic compounds and methods of using same
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11524963B2 (en) 2018-01-18 2022-12-13 Array Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11639346B2 (en) 2017-05-25 2023-05-02 Araxes Pharma Llc Quinazoline derivatives as modulators of mutant KRAS, HRAS or NRAS
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2019102203A (en) * 2012-07-11 2019-03-05 Блюпринт Медсинс Корпорейшн FIBROBLAST GROWTH FACTOR RECEPTOR INHIBITORS
US9499522B2 (en) 2013-03-15 2016-11-22 Blueprint Medicines Corporation Compositions useful for treating disorders related to kit
HUE059041T2 (en) 2013-10-17 2022-10-28 Blueprint Medicines Corp Process for preparing compositions useful for treating disorders related to kit
US9695165B2 (en) 2014-01-15 2017-07-04 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
WO2016107832A1 (en) 2014-12-30 2016-07-07 F. Hoffmann-La Roche Ag Novel tetrahydropyridopyrimidines and tetrahydropyridopyridines for the treatment and prophylaxis of hepatitis b virus infection
WO2016127074A1 (en) 2015-02-06 2016-08-11 Blueprint Medicines Corporation 2-(pyridin-3-yl)-pyrimidine derivatives as ret inhibitors
MX2018001032A (en) 2015-07-24 2018-11-09 Blueprint Medicines Corp Compounds useful for treating disorders related to kit and pdgfr.
JP6877407B2 (en) 2015-08-26 2021-05-26 ブループリント メディシンズ コーポレイション Compounds and compositions useful for the treatment of NTRK-related disorders
CN108473468B (en) 2015-11-02 2020-06-16 蓝图药品公司 Inhibitors of RET
AR106756A1 (en) 2015-11-19 2018-02-14 Blueprint Medicines Corp USEFUL COMPOUNDS AND COMPOSITIONS FOR THE TREATMENT OF NTRK-RELATED DISORDERS
TW201738228A (en) 2016-03-17 2017-11-01 藍圖醫藥公司 Inhibitors of RET
FI3442977T3 (en) 2016-04-15 2023-09-26 Blueprint Medicines Corp Inhibitors of activin receptor-like kinase
WO2018017983A1 (en) 2016-07-22 2018-01-25 Blueprint Medicines Corporation Compounds useful for treating disorders related to ret
WO2018022761A1 (en) 2016-07-27 2018-02-01 Blueprint Medicines Corporation Substituted cyclopentane-amides for treating disorders related to ret
WO2018183712A1 (en) 2017-03-31 2018-10-04 Blueprint Medicines Corporation Pyrrolo[1,2-b]pyridazine compounds and compositions useful for treating disorders related to kit and pdgfr
WO2019079649A1 (en) 2017-10-18 2019-04-25 Blueprint Medicines Corporation Substituted pyrrolopyridines as inhibitors of activin receptor-like kinase
MX2020010417A (en) 2018-04-03 2021-01-08 Blueprint Medicines Corp Ret inhibitor for use in treating cancer having a ret alteration.
CN117062809A (en) * 2021-03-26 2023-11-14 杭州普洛药物研究院有限公司 Bicyclic heterocyclic FGFR4 inhibitors, pharmaceutical compositions and formulations comprising the same and uses thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002076985A1 (en) * 2001-03-23 2002-10-03 Smithkline Beecham Corporation Compounds useful as kinase inhibitors for the treatment of hyperproliferative diseases
WO2006039718A2 (en) * 2004-10-01 2006-04-13 Amgen Inc Aryl nitrogen-containing bicyclic compounds and their use as kinase inhibitors
WO2010028236A1 (en) 2008-09-05 2010-03-11 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
WO2011034907A2 (en) 2009-09-16 2011-03-24 Avila Therapeutics, Inc. Protein kinase conjugates and inhibitors
WO2013179034A1 (en) * 2012-05-30 2013-12-05 Astex Therapeutics Limited New compounds
WO2014011900A2 (en) * 2012-07-11 2014-01-16 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor

Family Cites Families (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUT76853A (en) 1994-11-14 1997-12-29 Warner Lambert Co 6-aryl pyrido[2,3-d]pyrimidines and naphthyridines for inhibiting protein tyrosine kinase mediated cellular proliferation and pharmaceutical compositions containing the same
US6403799B1 (en) 1999-10-21 2002-06-11 Agouron Pharmaceuticals, Inc. Methods for the preparation of intermediates in the synthesis of HIV-protease inhibitors
EP1235815A1 (en) 1999-11-22 2002-09-04 Warner-Lambert Company Quinazolines and their use for inhibiting cyclin-dependent kinase enzymes
AU2001245401A1 (en) 2000-03-01 2001-09-12 Sumitomo Pharmaceuticals Company, Limited Hydrazones and analogs as cholesterol lowering agents
BR0112857A (en) 2000-08-04 2005-02-09 Warner Lambert Co 2- (4-Pyridyl) amino-6-dialkoxyphenyl-pyrido- [2,3-d] pyrimidin-7-ones compounds
GEP20063909B (en) 2002-01-22 2006-08-25 Warner Lambert Co 2-(PYRIDIN-2-YLAMINO)-PYRIDO[2,3d] PYRIMIDIN-7-ONES
US20050009849A1 (en) 2003-01-03 2005-01-13 Veach Darren R. Pyridopyrimidine kinase inhibitors
WO2005030131A2 (en) 2003-09-23 2005-04-07 Replidyne, Inc Bis-quinazoline compounds for the treatment of bacterial infections
CA2673003A1 (en) 2006-12-22 2008-07-03 Novartis Ag Quinazolines for pdk1 inhibition
WO2009046448A1 (en) 2007-10-04 2009-04-09 Intellikine, Inc. Chemical entities and therapeutic uses thereof
KR20110025224A (en) 2008-06-27 2011-03-09 아빌라 테라퓨틱스, 인크. Heteroaryl compounds and uses thereof
EP2342230A1 (en) 2008-09-03 2011-07-13 Licentia Ltd. Materials and methods for inhibiting cancer cell invasion related to fgfr4
EA019110B1 (en) 2008-12-29 2014-01-30 Фовеа Фармасьютикалз Substituted quinazoline compounds
US8829199B2 (en) 2009-08-07 2014-09-09 Chugai Seiyaku Kabushiki Kaisha Aminopyrazole derivative
UY33227A (en) 2010-02-19 2011-09-30 Novartis Ag PIRROLOPIRIMIDINE COMPOUNDS AS INHIBITORS OF THE CDK4 / 6
GB201007286D0 (en) 2010-04-30 2010-06-16 Astex Therapeutics Ltd New compounds
MX340408B (en) 2011-04-21 2016-07-07 Ionis Pharmaceuticals Inc Modulation of hepatitis b virus (hbv) expression.
KR102052670B1 (en) 2011-05-17 2019-12-06 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 Kinase inhibitors
FR2985257B1 (en) 2011-12-28 2014-02-14 Sanofi Sa FGFS RECEPTOR AGONISTIC DIMERS (FGFRS), PREPARATION METHOD AND THERAPEUTIC USE THEREOF
MY171055A (en) 2012-01-19 2019-09-23 Taiho Pharmaceutical Co Ltd 3,5-disubstituted alkynylbenzene compound and salt thereof
WO2014026125A1 (en) 2012-08-10 2014-02-13 Incyte Corporation Pyrazine derivatives as fgfr inhibitors
WO2014044846A1 (en) 2012-09-24 2014-03-27 Evotec (Uk) Ltd. 3-(aryl- or heteroaryl-amino)-7-(3,5-dimethoxyphenyl)isoquinoline derivatives as fgfr inhibitors useful for the treatment of proliferative disorders or dysplasia
TWI629266B (en) 2012-12-28 2018-07-11 藍印藥品公司 Inhibitors of the fibroblast growth factor receptor
HUE040434T2 (en) 2013-02-21 2019-03-28 Pfizer Solid forms of a selective cdk4/6 inhibitor
US9499522B2 (en) 2013-03-15 2016-11-22 Blueprint Medicines Corporation Compositions useful for treating disorders related to kit
AR095464A1 (en) 2013-03-15 2015-10-21 Celgene Avilomics Res Inc HETEROARILO COMPOUNDS AND USES OF THE SAME
JP6576325B2 (en) 2013-03-15 2019-09-18 セルジーン シーエーアール エルエルシー Heteroaryl compounds and their use
HUE059041T2 (en) 2013-10-17 2022-10-28 Blueprint Medicines Corp Process for preparing compositions useful for treating disorders related to kit
US9334263B2 (en) 2013-10-17 2016-05-10 Blueprint Medicines Corporation Compositions useful for treating disorders related to kit
LT3057943T (en) 2013-10-18 2018-11-12 Eisai R&D Management Co., Ltd. Pyrimidine fgfr4 inhibitors
KR101862493B1 (en) 2013-10-25 2018-05-29 노파르티스 아게 Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
RU2704112C2 (en) 2013-10-25 2019-10-24 Блюпринт Медсинс Корпорейшн Fibroblast growth factor receptor inhibitors
AU2015204763A1 (en) 2014-01-10 2016-07-21 Johnson & Johnson Consumer Inc. Process for making tablet using radiofrequency and lossy coated particles
US9695165B2 (en) 2014-01-15 2017-07-04 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
WO2016022569A1 (en) 2014-08-04 2016-02-11 Blueprint Medicines Corporation Compositions useful for treating disorders related to kit
WO2016064960A1 (en) 2014-10-22 2016-04-28 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
TWI712601B (en) 2015-02-20 2020-12-11 美商英塞特公司 Bicyclic heterocycles as fgfr inhibitors
MA41551A (en) 2015-02-20 2017-12-26 Incyte Corp BICYCLIC HETEROCYCLES USED AS FGFR4 INHIBITORS
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
MX2018001032A (en) 2015-07-24 2018-11-09 Blueprint Medicines Corp Compounds useful for treating disorders related to kit and pdgfr.
JP6877407B2 (en) 2015-08-26 2021-05-26 ブループリント メディシンズ コーポレイション Compounds and compositions useful for the treatment of NTRK-related disorders
EP3365335B1 (en) 2015-10-23 2024-02-14 Array Biopharma, Inc. 2-aryl- and 2-heteroaryl-substituted 2-pyridazin-3(2h)-one compounds as inhibitors of fgfr tyrosine kinases
CN108473468B (en) 2015-11-02 2020-06-16 蓝图药品公司 Inhibitors of RET
AR106756A1 (en) 2015-11-19 2018-02-14 Blueprint Medicines Corp USEFUL COMPOUNDS AND COMPOSITIONS FOR THE TREATMENT OF NTRK-RELATED DISORDERS
TW201738228A (en) 2016-03-17 2017-11-01 藍圖醫藥公司 Inhibitors of RET
FI3442977T3 (en) 2016-04-15 2023-09-26 Blueprint Medicines Corp Inhibitors of activin receptor-like kinase

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002076985A1 (en) * 2001-03-23 2002-10-03 Smithkline Beecham Corporation Compounds useful as kinase inhibitors for the treatment of hyperproliferative diseases
WO2006039718A2 (en) * 2004-10-01 2006-04-13 Amgen Inc Aryl nitrogen-containing bicyclic compounds and their use as kinase inhibitors
WO2010028236A1 (en) 2008-09-05 2010-03-11 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
WO2011034907A2 (en) 2009-09-16 2011-03-24 Avila Therapeutics, Inc. Protein kinase conjugates and inhibitors
WO2013179034A1 (en) * 2012-05-30 2013-12-05 Astex Therapeutics Limited New compounds
WO2014011900A2 (en) * 2012-07-11 2014-01-16 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
BROWN, AP ET AL., TOXICOL. PATHOL., 2005, pages 449 - 455
CAO ET AL., CANCER RES, vol. 70, no. 16, 2010, pages 6497 - 6508
DING ET AL., NATURE, vol. 455, no. 7216, 2008, pages 1069 - 1075
HO ET AL., JOURNAL OF HEPATOLOGY, vol. 50, 2009, pages 118 - 27
ROIDL ET AL., ONCOGENE, vol. 29, no. 10, 2010, pages 1543 - 1552
SAWEY ET AL., CANCER CELL, vol. 19, 2011, pages 347 - 358
TAYLOR ET AL., J CLIN INVEST, vol. 119, 2009, pages 3395 - 3407
VERGNES ET AL., CELL METABOLISM, vol. 17, 2013, pages 916 - 28
WU ET AL., J BIOL CHEM, vol. 285, no. 8, 2010, pages 5165 - 5170
WUTS; GREENE: "Protective Groups in Organic Synthesis, 4th ed.", 2006, JOHN WILEY & SONS
ZAID ET AL., CLIN. CANCER RES., 2013, pages 809

Cited By (178)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10758542B2 (en) 2009-07-09 2020-09-01 Array Biopharma Inc. Substituted pyrazolo[l,5-a]pyrimidine compounds as Trk kinase inhibitors
US10251889B2 (en) 2009-07-09 2019-04-09 Array BioPharm Inc. Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US10273207B2 (en) 2013-03-15 2019-04-30 Araxes Pharma Llc Covalent inhibitors of kras G12C
US10618902B2 (en) 2013-03-15 2020-04-14 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10774052B2 (en) 2013-03-15 2020-09-15 Celgene Car Llc Heteroaryl compounds and uses thereof
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US9695132B2 (en) 2013-03-15 2017-07-04 Celgene Car Llc Heteroaryl compounds and uses thereof
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US10919850B2 (en) 2013-03-15 2021-02-16 Araxes Pharma Llc Covalent inhibitors of KRas G12C
US10189794B2 (en) 2013-03-15 2019-01-29 Celgene Car Llc Heteroaryl compounds and uses thereof
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10370386B2 (en) 2013-10-10 2019-08-06 Araxes Pharma Llc Substituted quinolines as inhibitors of KRAS G12C
US10927125B2 (en) 2013-10-10 2021-02-23 Araxes Pharma Llc Substituted cinnolines as inhibitors of KRAS G12C
US11878985B2 (en) 2013-10-10 2024-01-23 Araxes Pharma Llc Substituted quinazolines as inhibitors of KRAS G12C
US10912774B2 (en) 2013-10-18 2021-02-09 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US10537571B2 (en) 2013-10-18 2020-01-21 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US9434697B2 (en) 2013-10-18 2016-09-06 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US9730931B2 (en) 2013-10-18 2017-08-15 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US10221154B2 (en) 2013-10-25 2019-03-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10875837B2 (en) 2013-10-25 2020-12-29 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10111874B2 (en) 2014-09-18 2018-10-30 Araxes Pharma Llc Combination therapies for treatment of cancer
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
JP2020147592A (en) * 2015-02-20 2020-09-17 インサイト・コーポレイションIncyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2016134314A1 (en) * 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
JP2018507214A (en) * 2015-02-20 2018-03-15 インサイト・コーポレイションIncyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2016134294A1 (en) * 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
US10246424B2 (en) 2015-04-10 2019-04-02 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10829458B2 (en) 2015-04-10 2020-11-10 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10562888B2 (en) 2015-04-14 2020-02-18 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
US11498916B2 (en) 2015-04-14 2022-11-15 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
US10428064B2 (en) 2015-04-15 2019-10-01 Araxes Pharma Llc Fused-tricyclic inhibitors of KRAS and methods of use thereof
US10174028B2 (en) 2015-07-16 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10174027B2 (en) 2015-07-16 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
WO2017011776A1 (en) 2015-07-16 2017-01-19 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10138243B2 (en) 2015-07-16 2018-11-27 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10023570B2 (en) 2015-07-16 2018-07-17 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10144724B2 (en) 2015-07-22 2018-12-04 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10351550B2 (en) 2015-07-22 2019-07-16 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10975071B2 (en) 2015-09-28 2021-04-13 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10647703B2 (en) 2015-09-28 2020-05-12 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10882847B2 (en) 2015-09-28 2021-01-05 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10689356B2 (en) 2015-09-28 2020-06-23 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10875842B2 (en) 2015-09-28 2020-12-29 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10858343B2 (en) 2015-09-28 2020-12-08 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10730867B2 (en) 2015-09-28 2020-08-04 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10907215B2 (en) 2015-10-26 2021-02-02 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10724102B2 (en) 2015-10-26 2020-07-28 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US11021470B2 (en) 2015-11-16 2021-06-01 Araxes Pharma Llc 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
US10414757B2 (en) 2015-11-16 2019-09-17 Araxes Pharma Llc 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
WO2017176744A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Methods of treating pediatric cancers
US10137127B2 (en) 2016-04-04 2018-11-27 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10588908B2 (en) 2016-04-04 2020-03-17 Loxo Oncology, Inc. Methods of treating pediatric cancers
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
WO2017176751A1 (en) 2016-04-04 2017-10-12 Loxo Oncology, Inc. Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US11484535B2 (en) 2016-04-04 2022-11-01 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a] pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10668072B2 (en) 2016-04-04 2020-06-02 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US11191766B2 (en) 2016-04-04 2021-12-07 Loxo Oncology, Inc. Methods of treating pediatric cancers
US11357769B2 (en) 2016-05-10 2022-06-14 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
US11214571B2 (en) 2016-05-18 2022-01-04 Array Biopharma Inc. Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
US10646488B2 (en) 2016-07-13 2020-05-12 Araxes Pharma Llc Conjugates of cereblon binding compounds and G12C mutant KRAS, HRAS or NRAS protein modulating compounds and methods of use thereof
CN107793395A (en) * 2016-09-01 2018-03-13 南京药捷安康生物科技有限公司 Fibroblast growth factor acceptor inhibitor and application thereof
WO2018040885A1 (en) * 2016-09-01 2018-03-08 南京药捷安康生物科技有限公司 Inhibitors of fibroblast growth factor receptor and use thereof
CN107793395B (en) * 2016-09-01 2021-01-05 南京药捷安康生物科技有限公司 Fibroblast growth factor receptor inhibitors and uses thereof
RU2732127C1 (en) * 2016-09-01 2020-09-11 Наньцзин Транстера Биосайенсиз Ко. Лтд. Fibroblast growth factor receptor inhibitors and use thereof
US10744139B2 (en) 2016-09-01 2020-08-18 Nanjing Transthera Biosciences Co. Ltd. Inhibitors of fibroblast growth factor receptor and use thereof
JP2019529374A (en) * 2016-09-01 2019-10-17 南京薬捷安康生物科技有限公司 Inhibitors of fibroblast growth factor receptor and their use
WO2018049233A1 (en) 2016-09-08 2018-03-15 Nicolas Stransky Inhibitors of the fibroblast growth factor receptor in combination with cyclin-dependent kinase inhibitors
US10280172B2 (en) 2016-09-29 2019-05-07 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10723738B2 (en) 2016-09-29 2020-07-28 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
US10377743B2 (en) 2016-10-07 2019-08-13 Araxes Pharma Llc Inhibitors of RAS and methods of use thereof
US10172845B2 (en) 2016-10-10 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US11648243B2 (en) 2016-10-10 2023-05-16 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10144734B2 (en) 2016-10-10 2018-12-04 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10172851B2 (en) 2016-10-10 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10555944B2 (en) 2016-10-10 2020-02-11 Eli Lilly And Company Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10137124B2 (en) 2016-10-10 2018-11-27 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10112942B2 (en) 2016-10-10 2018-10-30 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
EP3753939A1 (en) 2016-10-10 2020-12-23 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10881652B2 (en) 2016-10-10 2021-01-05 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
EP4144735A1 (en) 2016-10-10 2023-03-08 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10441581B2 (en) 2016-10-10 2019-10-15 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10953005B1 (en) 2016-10-10 2021-03-23 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
WO2018071454A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018071447A1 (en) 2016-10-10 2018-04-19 Andrews Steven W Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US11091486B2 (en) 2016-10-26 2021-08-17 Array Biopharma, Inc Process for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
US11555036B2 (en) 2016-12-19 2023-01-17 Abbisko Therapeutics Co., Ltd. FGFR4 inhibitor, preparation method therefor and pharmaceutical use thereof
EP3524603A4 (en) * 2016-12-19 2020-04-22 Abbisko Therapeutics Co., Ltd. Fgfr4 inhibitor, preparation method therefor and pharmaceutical use thereof
US10968220B2 (en) 2016-12-19 2021-04-06 Abbisko Therapeutics Co., Ltd. FGFR4 inhibitor, preparation method therefor and pharmaceutical use thereof
WO2018136661A1 (en) 2017-01-18 2018-07-26 Andrews Steven W SUBSTITUTED PYRAZOLO[1,5-a]PYRAZINE COMPOUNDS AS RET KINASE INHIBITORS
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
US11168090B2 (en) 2017-01-18 2021-11-09 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyrazines as RET kinase inhibitors
US11851434B2 (en) 2017-01-18 2023-12-26 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyrazine compounds as ret kinase inhibitors
US11136308B2 (en) 2017-01-26 2021-10-05 Araxes Pharma Llc Substituted quinazoline and quinazolinone compounds and methods of use thereof
US11059819B2 (en) 2017-01-26 2021-07-13 Janssen Biotech, Inc. Fused hetero-hetero bicyclic compounds and methods of use thereof
WO2018140514A1 (en) * 2017-01-26 2018-08-02 Araxes Pharma Llc 1-(6-(3-hydroxynaphthalen-1-yl)quinazolin-2-yl)azetidin-1-yl)prop-2-en-1-one derivatives and similar compounds as kras g12c inhibitors for the treatment of cancer
US11274093B2 (en) 2017-01-26 2022-03-15 Araxes Pharma Llc Fused bicyclic benzoheteroaromatic compounds and methods of use thereof
US11279689B2 (en) 2017-01-26 2022-03-22 Araxes Pharma Llc 1-(3-(6-(3-hydroxynaphthalen-1-yl)benzofuran-2-yl)azetidin-1 yl)prop-2-en-1-one derivatives and similar compounds as KRAS G12C modulators for treating cancer
US11358959B2 (en) 2017-01-26 2022-06-14 Araxes Pharma Llc Benzothiophene and benzothiazole compounds and methods of use thereof
US10688100B2 (en) 2017-03-16 2020-06-23 Array Biopharma Inc. Macrocylic compounds as ROS1 kinase inhibitors
US10966985B2 (en) 2017-03-16 2021-04-06 Array Biopharma Inc. Macrocyclic compounds as ROS1 kinase inhibitors
WO2018170381A1 (en) 2017-03-16 2018-09-20 Andrews Steven W Macrocyclic compounds as ros1 kinase inhibitors
CN108864081A (en) * 2017-05-09 2018-11-23 南京圣和药业股份有限公司 Fgfr4 inhibitor and its preparation and application
CN108864081B (en) * 2017-05-09 2021-11-23 南京圣和药业股份有限公司 FGFR4 inhibitor and preparation and application thereof
WO2018205916A1 (en) * 2017-05-09 2018-11-15 南京圣和药业股份有限公司 Fgfr4 inhibitor and preparation and use thereof
US10745385B2 (en) 2017-05-25 2020-08-18 Araxes Pharma Llc Covalent inhibitors of KRAS
US11639346B2 (en) 2017-05-25 2023-05-02 Araxes Pharma Llc Quinazoline derivatives as modulators of mutant KRAS, HRAS or NRAS
US11377441B2 (en) 2017-05-25 2022-07-05 Araxes Pharma Llc Covalent inhibitors of KRAS
US10736897B2 (en) 2017-05-25 2020-08-11 Araxes Pharma Llc Compounds and methods of use thereof for treatment of cancer
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
WO2019075108A1 (en) 2017-10-10 2019-04-18 Metcalf Andrew T Crystalline forms
WO2019075114A1 (en) 2017-10-10 2019-04-18 Mark Reynolds Formulations comprising 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazab icyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
CN111278823B (en) * 2017-10-24 2023-01-24 上海新契博生物科技有限公司 Heterocyclic compounds as fibroblast growth factor receptor inhibitors
WO2019080878A1 (en) * 2017-10-24 2019-05-02 Advent Pharma Co. Ltd. Heterocyclics as inhibitors of fibroblast growth factor receptor
CN111278823A (en) * 2017-10-24 2020-06-12 上海新契博生物科技有限公司 Heterocyclic compounds as fibroblast growth factor receptor inhibitors
CN109745325A (en) * 2017-11-08 2019-05-14 上海翰森生物医药科技有限公司 FGFR4 inhibitor, preparation method and use
US11603374B2 (en) 2018-01-18 2023-03-14 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
US11524963B2 (en) 2018-01-18 2022-12-13 Array Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
WO2019143994A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
WO2019143977A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
US11472802B2 (en) 2018-01-18 2022-10-18 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridine compounds as RET kinase inhibitors
CN110092798A (en) * 2018-01-29 2019-08-06 上海新契博生物科技有限公司 It is a kind of as the heterocyclic compound and its synthetic method of FGFR inhibitor and application
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
CN110577524B (en) * 2018-06-07 2022-01-28 北京大学深圳研究生院 Kinase selective inhibitor
CN110577524A (en) * 2018-06-07 2019-12-17 北京大学深圳研究生院 Kinase selective inhibitor
WO2019233438A1 (en) * 2018-06-07 2019-12-12 北京大学深圳研究生院 Kinase selective inhibitor
WO2020028258A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide
WO2020055672A1 (en) 2018-09-10 2020-03-19 Array Biopharma Inc. Fused heterocyclic compounds as ret kinase inhibitors
KR102531596B1 (en) * 2018-09-14 2023-05-11 아비스코 테라퓨틱스 컴퍼니 리미티드 FGFR inhibitor, manufacturing method and use thereof
KR20210019510A (en) * 2018-09-14 2021-02-22 아비스코 테라퓨틱스 컴퍼니 리미티드 FGFR inhibitor, its manufacturing method and use
JP7152794B2 (en) 2018-09-14 2022-10-13 アビスコ セラピューティクス カンパニー リミテッド FGFR inhibitor, production method and application thereof
JP2021527678A (en) * 2018-09-14 2021-10-14 アビスコ セラピューティクス カンパニー リミテッド FGFR inhibitor, its manufacturing method and application
CN111138459A (en) * 2018-11-06 2020-05-12 南京圣和药业股份有限公司 Optical isomer of FGFR4 inhibitor and application thereof
WO2020119606A1 (en) * 2018-12-10 2020-06-18 Guangdong Newopp Biopharmaceuticals Co., Ltd. Heterocyclic compounds as inhibitors of fibroblast growth factor receptor
WO2020131627A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
CN111902400A (en) * 2019-03-05 2020-11-06 保仕健生物科技(上海)有限公司 Aromatic derivatives, process for their preparation and their medical use
WO2020177534A1 (en) * 2019-03-05 2020-09-10 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof
JP2022523448A (en) * 2019-03-08 2022-04-22 ショウヤオ ホールディングス(ベイジン) カンパニー, リミテッド FGFR4 kinase inhibitor, its production method and use
JP7378488B2 (en) 2019-03-08 2023-11-13 ショウヤオ ホールディングス(ベイジン) カンパニー, リミテッド FGFR4 kinase inhibitor, its production method and use
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
WO2021134004A1 (en) 2019-12-27 2021-07-01 Schrodinger, Inc. Cyclic compounds and methods of using same
WO2022055963A1 (en) 2020-09-10 2022-03-17 Schrödinger, Inc. Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer
WO2022087433A1 (en) * 2020-10-23 2022-04-28 Dana-Farber Cancer Institute, Inc. Covalent inhibitors of creatine kinase (ck) and uses thereof for treating and preventing cancer
WO2022164789A1 (en) 2021-01-26 2022-08-04 Schrödinger, Inc. Tricyclic compounds useful in the treatment of cancer, autoimmune and inflammatory disoders
WO2022197898A1 (en) 2021-03-18 2022-09-22 Schrödinger, Inc. Cyclic compounds and methods of using same
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
US20150197519A1 (en) 2015-07-16
US20170253593A1 (en) 2017-09-07
US10000490B2 (en) 2018-06-19
US9695165B2 (en) 2017-07-04

Similar Documents

Publication Publication Date Title
US10000490B2 (en) Inhibitors of the fibroblast growth factor receptor
US10875837B2 (en) Inhibitors of the fibroblast growth factor receptor
RU2679130C2 (en) Inhibitors of fibroblast growth factor receptor
TWI629266B (en) Inhibitors of the fibroblast growth factor receptor
EP2978759B1 (en) Benzimidazolone derivatives as bromodomain inhibitors
JP4324338B2 (en) Condensed imidazole compound and antidiabetic agent
CN111635408B (en) Triazolo-pyrimidine compounds and uses thereof
Peng et al. Discovery of 2-(2-aminopyrimidin-5-yl)-4-morpholino-N-(pyridin-3-yl) quinazolin-7-amines as novel PI3K/mTOR inhibitors and anticancer agents
CN110938071A (en) Heteroaromatic compound with kinase inhibition activity
CN116063324A (en) Inhibitors of BTK and/or RET having macrocyclic structure

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15703351

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 23.11.2016)

122 Ep: pct application non-entry in european phase

Ref document number: 15703351

Country of ref document: EP

Kind code of ref document: A1